1
|
Zhou L, Zhu JQ, Kou JT, Xu WL, Lyu SC, Du GS, Yang HW, Wang JF, Hu XP, Yu CZ, Yuan CH, Han DD, Sang CQ, Li B, Gao J, Qi HZ, Wang LM, Lyu L, Liu H, Wu JY, Lang R, He Q, Li XL. Chinese expert consensus on quantitatively monitoring and assessing immune cell function status and its clinical application (2024 edition). Hepatobiliary Pancreat Dis Int 2024:S1499-3872(24)00125-5. [PMID: 39448347 DOI: 10.1016/j.hbpd.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Affiliation(s)
- Lin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Ji-Qiao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jian-Tao Kou
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wen-Li Xu
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Shao-Cheng Lyu
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Guo-Sheng Du
- Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang 110010, China
| | - Hong-Wei Yang
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang 110010, China
| | - Jian-Feng Wang
- Department of Interventional Therapy, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiao-Peng Hu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chun-Zhao Yu
- Department of General Surgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Chun-Hui Yuan
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Dong-Dong Han
- Liver Transplantation Department, China-Japan Friendship Hospital, Beijing 100029, China
| | - Cui-Qin Sang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Bo Li
- Department of Hepatobiliary Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100871, China
| | - Hai-Zhi Qi
- Department of General Surgery/Organ Transplant Center, The Second Xiang Ya Hospital of Central South Univercity, Changsha 410011, China
| | - Li-Ming Wang
- Organ Transplant Center, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Ling Lyu
- Department of General Surgery, Jiangsu Provincial People's Hospital, Nanjing 210029, China
| | - Hao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Jian-Yong Wu
- Kidney Transplant Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ren Lang
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Qiang He
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Xian-Liang Li
- Department of Hepatobiliary and Pancreatic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
2
|
Kori M, Kasavi C, Arga KY. Exploring COVID-19 Pandemic Disparities with Transcriptomic Meta-analysis from the Perspective of Personalized Medicine. J Microbiol 2024; 62:785-798. [PMID: 38980578 PMCID: PMC11436439 DOI: 10.1007/s12275-024-00154-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Infection with SARS-CoV2, which is responsible for COVID-19, can lead to differences in disease development, severity and mortality rates depending on gender, age or the presence of certain diseases. Considering that existing studies ignore these differences, this study aims to uncover potential differences attributable to gender, age and source of sampling as well as viral load using bioinformatics and multi-omics approaches. Differential gene expression analyses were used to analyse the phenotypic differences between SARS-CoV-2 patients and controls at the mRNA level. Pathway enrichment analyses were performed at the gene set level to identify the activated pathways corresponding to the differences in the samples. Drug repurposing analysis was performed at the protein level, focusing on host-mediated drug candidates to uncover potential therapeutic differences. Significant differences (i.e. the number of differentially expressed genes and their characteristics) were observed for COVID-19 at the mRNA level depending on the sample source, gender and age of the samples. The results of the pathway enrichment show that SARS-CoV-2 can be combated more effectively in the respiratory tract than in the blood samples. Taking into account the different sample sources and their characteristics, different drug candidates were identified. Evaluating disease prediction, prevention and/or treatment strategies from a personalised perspective is crucial. In this study, we not only evaluated the differences in COVID-19 from a personalised perspective, but also provided valuable data for further experimental and clinical efforts. Our findings could shed light on potential pandemics.
Collapse
Affiliation(s)
- Medi Kori
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752, Istanbul, Turkey.
- Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752, Istanbul, Turkey.
| | - Ceyda Kasavi
- Department of Bioengineering, Marmara University, 34722, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722, Istanbul, Turkey
- Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34722, Istanbul, Turkey
| |
Collapse
|
3
|
Shrwani KJ, Mahallawi WH, Mohana AI, Algaissi A, Dhayhi N, Sharwani NJ, Gadour E, Aldossari SM, Asiri H, Kameli N, Asiri AY, Asiri AM, Sherwani AJ, Cunliffe N, Zhang Q. Mucosal immunity in upper and lower respiratory tract to MERS-CoV. Front Immunol 2024; 15:1358885. [PMID: 39281686 PMCID: PMC11392799 DOI: 10.3389/fimmu.2024.1358885] [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: 12/20/2023] [Accepted: 07/15/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Middle East respiratory syndrome coronavirus (MERS-CoV) has emerged as a deadly pathogen with a mortality rate of up to 36.2%. MERS-CoV can cause severe respiratory tract disease and multiorgan failure. Therefore, therapeutic vaccines are urgently needed. This intensive review explores the human immune responses and their immunological mechanisms during MERS-CoV infection in the mucosa of the upper and lower respiratory tracts (URT and LRT, respectively). Objective The aim of this study is to provide a valuable, informative, and critical summary of the protective immune mechanisms against MERS-CoV infection in the URT/LRT for the purpose of preventing and controlling MERS-CoV disease and designing effective therapeutic vaccines. Methods In this review, we focus on the immune potential of the respiratory tract following MERS-CoV infection. We searched PubMed, Embase, Web of Science, Cochrane, Scopus, and Google Scholar using the following terms: "MERS-CoV", "B cells", "T cells", "cytokines", "chemokines", "cytotoxic", and "upper and lower respiratory tracts". Results We found and included 152 studies in this review. We report that the cellular innate immune response, including macrophages, dendritic cells, and natural killer cells, produces antiviral substances such as interferons and interleukins to prevent the virus from spreading. In the adaptive and humoral immune responses, CD4+ helper T cells, CD8+ cytotoxic T cells, B cells, and plasma cells protect against MERS-CoV infection in URT and LRT. Conclusion The human nasopharynx-associated lymphoid tissue (NALT) and bronchus-associated lymphoid tissue (BALT) could successfully limit the spread of several respiratory pathogens. However, in the case of MERS-CoV infection, limited research has been conducted in humans with regard to immunopathogenesis and mucosal immune responses due to the lack of relevant tissues. A better understanding of the immune mechanisms of the URT and LRT is vital for the design and development of effective MERS-CoV vaccines.
Collapse
Affiliation(s)
- Khalid J Shrwani
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Public Health Authority, Saudi Center for Disease Prevention and Control (SCDC), Jazan, Saudi Arabia
| | - Waleed H Mahallawi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Abdulrhman I Mohana
- Department of Antimicrobial Resistance, Public Health Authority, Riyadh, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Emerging and Endemic Infectious Diseases Research Unit, Health Sciences Research Center, Jazan University, Jazan, Saudi Arabia
| | - Nabil Dhayhi
- Department of Pediatrics, King Fahad Central Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nouf J Sharwani
- Department of Surgery, Mohammed bin Nasser Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Eyad Gadour
- Department of Gastroenterology and Hepatology, King Abdulaziz National Guard Hospital, Ahsa, Saudi Arabia
- Department of Medicine, Faculty of Medicine, Zamzam University College, Khartoum, Sudan
| | - Saeed M Aldossari
- Medical Laboratory Technology Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Hasan Asiri
- Medical Laboratory Department, Prince Mohammed bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Nader Kameli
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ayad Y Asiri
- Intensive Care Unit Department, Al Inma Medical Group, Al Hayat National Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah M Asiri
- Preventive Medicine Assistant Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Alaa J Sherwani
- Department of Pediatrics, Abu-Arish General Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nigel Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Qibo Zhang
- Academic and Research Departments, Section of Immunology, School of Biosciences, University of Surrey, Surrey, United Kingdom
| |
Collapse
|
4
|
Iuliano M, Mongiovì RM, Parente A, Kertusha B, Carraro A, Marocco R, Mancarella G, Del Borgo C, Fondaco L, Grimaldi L, Dorrucci M, Lichtner M, Mangino G, Romeo G. Dysregulated Inflammatory Cytokine Levels May Be Useful Markers in a Better Up-Dated Management of COVID-19. Curr Issues Mol Biol 2024; 46:8890-8902. [PMID: 39194742 DOI: 10.3390/cimb46080525] [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: 07/16/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infection characterized by the dysregulation of systemic cytokine levels. The measurement of serum levels of inflammatory cyto-/chemokines has been suggested as a tool in the management of COVID-19. The aim of this study is to highlight the significance of measured levels of interleukin (IL)-1α, IL-1β, IL-6, IL-8, IL-10, IL-12(p70), IL-27, interferon (IFN)γ, interferon gamma-induced protein (IP)-10, monocyte chemoattractant protein (MCP)-1, and tumor necrosis factor (TNF)-α in serum samples from infected and recovered subjects, possibly predictive of severity and/or duration of the disease. Serum samples from healthy (HD), positive at hospital admittance (T0), and recovered subjects (T1, 31-60, or 70-200 days post-negativization) were collected and tested through a bead-based cytometric assay and confirmed through ELISA. IL-10 levels were increased in the T0 group compared to both HD and T1. IL-27 significantly decreased in the 31-60 group. IL-1β significantly increased in the 70-200 day group. TNF-α significantly decreased in T0 compared to HD and in the 31-60 group versus HD. IP-10 significantly increased in T0 compared to HD. These results suggest that IP-10 could represent an early marker of clinical worsening, whereas IL-10 might be indicative of the possible onset of post-COVID-19 long syndrome.
Collapse
Affiliation(s)
- Marco Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Roberta Maria Mongiovì
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98122 Messina, Italy
| | - Alberico Parente
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Blerta Kertusha
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Anna Carraro
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Raffaella Marocco
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Giulia Mancarella
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Cosmo Del Borgo
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Laura Fondaco
- Department of Public Health and Infectious Disease, S. Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Lorenzo Grimaldi
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Maria Dorrucci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Miriam Lichtner
- Department of General Surgery and Surgical Specialty, Sapienza University of Rome, 00168 Rome, Italy
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| |
Collapse
|
5
|
Alqahtani SAM. Mucosal immunity in COVID-19: a comprehensive review. Front Immunol 2024; 15:1433452. [PMID: 39206184 PMCID: PMC11349522 DOI: 10.3389/fimmu.2024.1433452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Mucosal immunity plays a crucial role in defending against coronaviruses, particularly at respiratory sites, serving as the first line of defense against viral invasion and replication. Coronaviruses have developed various immune evasion strategies at the mucosal immune system, hindering the recognition of infected cells and evading antibody responses. Understanding the immune mechanisms and responses is crucial for developing effective vaccines and therapeutics against coronaviruses. The role of mucosal immunity in COVID-19 is significant, influencing both local and systemic immune responses to the virus. Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Mucosally delivered vaccines and those under clinical trials are being compared and contrasted to understand their effectiveness in inducing mucosal immunity against coronaviruses. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community-acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality. This comprehensive review article outlines the current evidence about the role of mucosal immune responses in the clearance of SARS-CoV-2 infection, as well as potential mucosal mechanisms of protection against (re-)infection. It also proposes that there is a significant role for mucosal immunity and for secretory as well as circulating IgA antibodies in COVID-19, and that it is important to elucidate this in order to comprehend especially the asymptomatic and mild states of the infection, which appear to account for the majority of cases. Moreover, it is possible that mucosal immunity can be exploited for beneficial diagnostic, therapeutic, or prophylactic purposes. The findings from recent studies on mucosal immunity in COVID-19 can be used to develop effective vaccines and treatments that can effectively target both mucosal and systemic immune responses.
Collapse
|
6
|
Du X, Yang X, Zhao J, Zhang J, Yu J, Ma L, Zhang W, Cen S, Ren X, He X. Design of novel broad-spectrum antiviral nucleoside analogues using natural bases ring-opening strategy. Drug Dev Res 2024; 85:e22237. [PMID: 39032059 DOI: 10.1002/ddr.22237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 07/22/2024]
Abstract
The global prevalence of RNA virus infections has presented significant challenges to public health in recent years, necessitating the expansion of its alternative therapeutic library. Due to its evolutional conservation, RNA-dependent RNA polymerase (RdRp) has emerged as a potential target for broad-spectrum antiviral nucleoside analogues. However, after over half a century of structural modification, exploring unclaimed chemical space using frequently-used structural substitution methods to design new nucleoside analogues is challenging. In this study, we explore the use of the "ring-opening" strategy to design new base mimics, thereby using these base mimics to design new nucleoside analogues with broad-spectrum antiviral activities. A total of 29 compounds were synthesized. Their activity against viral RdRp was initially screened using an influenza A virus RdRp high-throughput screening model. Then, the antiviral activity of 38a was verified against influenza virus strain A/PR/8/34 (H1N1), demonstrating a 50% inhibitory concentration (IC50) value of 9.95 μM, which was superior to that of ribavirin (the positive control, IC50 = 11.43 μM). Moreover, 38a also has inhibitory activity against coronavirus 229E with an IC50 of 30.82 μM. In addition, compounds 42 and 46f exhibit an 82% inhibition rate against vesicular stomatitis virus at a concentration of 20 μM and hardly induce cytotoxicity in host cells. This work demonstrates the feasibility of designing nucleoside analogues with "ring-opening" bases and suggests the "ring-opening" nucleosides may have greater polarity, and designing prodrugs is an important aspect of optimizing their antiviral activity. Future research should focus on enhancing the conformational restriction of open-loop bases to mimic Watson-Crick base pairing better and improve antiviral activity.
Collapse
Affiliation(s)
- Xingyi Du
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xingxing Yang
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jianyuan Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Jinyan Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiahui Yu
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Weina Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Xuhong Ren
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinhua He
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
7
|
El-Gendy ZA, Abdelazeem S, Abdel Jaleel GA, Ali ME, Mohamed A, Salah A, Raslan MA. Anti-inflammatory and anti-rheumatic effects of Phoenix dactylifera L. (date palm) seed by controlling cytokines and inhibiting JAK1/STAT3 pathway on CFA-induced arthritis rat and its phytochemical profiling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118138. [PMID: 38565410 DOI: 10.1016/j.jep.2024.118138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phoenix dactylifera L. (date palm) seed is widely used in Arabian traditional medicine to alleviate several health problems including inflammatory conditions. The herbal tea of date palm seed has been consumed by rheumatoid patients to relief their symptoms. AIM OF THE STUDY The purpose of this study was to investigate the claimed beneficial use of P. dactylifera L. (Sewy variety) seed (PDS) in the treatment of rheumatoid arthritis (RA) and its mechanism of action as well as to study its phytoconstituents. MATERIALS AND METHODS The anti-inflammatory and anti-oxidative properties of the non-polar and the polar extracts of PDS were studied using Complete Freund's adjuvant (CFA)-induced arthritis rat model. Paw edema, body weight, total nitrate/nitrite NOX content and cytokine markers were evaluated to monitor the progress of arthritis. Also, histological examination and thermal analysis were conducted. The phytoconstituent profiles of non-polar and polar extracts of PDS were investigated using liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). The multiple reactions monitoring mode (MRM) of liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) was used to quantify phenolic phytoconstituents in both extracts. RESULTS According to the findings, the polar and non-polar PDS extracts kept body weight comparable to those of healthy individuals while considerably lowering paw swelling, edema, and neutrophil infiltration. It also reduced the levels of Nuclear Factor Kappa B (NF-κB), Tumor Necrosis Factor Alpha (TNF-α), Interleukin 22, Interleukin 23, Interferon (IFN), Interleukin 17, Interleukin 1β, Interleukin 6, Interleukin 36, Janus Kinase 1 (JAK1), and Signal Transducer and Activator of Transcription 3 (STAT3). They also reduced the degenerative alterations caused by RA. Thermal research gave additional support for these findings. 83 phytoconstituents were identified in the non-polar PDS extract and 86 phytoconstituents were identified in the polar PDS extract. 74 of the identified phytoconstituents were common in both extracts. 33 phytoconstituents were identified here from P. dactylifera for the first time as far as we know. In MRM-LC-ESI-MS/MS analysis, the major phenolics in both extracts were chlorogenic acid, naringenin, and vanillin. Catechin was only detected in the non-polar PDS extract. On the other hand, apigenin, kaempferol, and hesperetin were only detected in the polar PDS extract. Generally, the polar PDS extract showed higher concentrations of the identified phenolics than the non-polar extract. CONCLUSIONS The PDS extracts especially the non-polar extract showed significant anti-inflammatory and anti-oxidative properties in the CFA-induced arthritis rat model. PDS might be used to produce RA medicines.
Collapse
Affiliation(s)
- Zeinab A El-Gendy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt.
| | - Shimaa Abdelazeem
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| | - Gehad A Abdel Jaleel
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt.
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Asmaa Mohamed
- Department of Computer Science, High Institute for Management Sciences, Belqas, Egypt.
| | - Ahmad Salah
- Department of Information Technology, College of Computing and Information Sciences, University of Technology and Applied Sciences, Ibri, Sultanate of Oman; Department of Computer Science, Faculty of Computers and Informatics, Zagazig University, Egypt.
| | - Mona A Raslan
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, 12622, Giza, Egypt.
| |
Collapse
|
8
|
Khodadadi Yazdi M, Seidi F, Hejna A, Zarrintaj P, Rabiee N, Kucinska-Lipka J, Saeb MR, Bencherif SA. Tailor-Made Polysaccharides for Biomedical Applications. ACS APPLIED BIO MATERIALS 2024; 7:4193-4230. [PMID: 38958361 PMCID: PMC11253104 DOI: 10.1021/acsabm.3c01199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Polysaccharides (PSAs) are carbohydrate-based macromolecules widely used in the biomedical field, either in their pure form or in blends/nanocomposites with other materials. The relationship between structure, properties, and functions has inspired scientists to design multifunctional PSAs for various biomedical applications by incorporating unique molecular structures and targeted bulk properties. Multiple strategies, such as conjugation, grafting, cross-linking, and functionalization, have been explored to control their mechanical properties, electrical conductivity, hydrophilicity, degradability, rheological features, and stimuli-responsiveness. For instance, custom-made PSAs are known for their worldwide biomedical applications in tissue engineering, drug/gene delivery, and regenerative medicine. Furthermore, the remarkable advancements in supramolecular engineering and chemistry have paved the way for mission-oriented biomaterial synthesis and the fabrication of customized biomaterials. These materials can synergistically combine the benefits of biology and chemistry to tackle important biomedical questions. Herein, we categorize and summarize PSAs based on their synthesis methods, and explore the main strategies used to customize their chemical structures. We then highlight various properties of PSAs using practical examples. Lastly, we thoroughly describe the biomedical applications of tailor-made PSAs, along with their current existing challenges and potential future directions.
Collapse
Affiliation(s)
- Mohsen Khodadadi Yazdi
- Division
of Electrochemistry and Surface Physical Chemistry, Faculty of Applied
Physics and Mathematics, Gdańsk University
of Technology, Narutowicza
11/12, 80-233 Gdańsk, Poland
- Advanced
Materials Center, Gdańsk University
of Technology, Narutowicza
11/12, 80-233 Gdańsk, Poland
| | - Farzad Seidi
- Jiangsu
Co−Innovation Center for Efficient Processing and Utilization
of Forest Resources and International Innovation Center for Forest
Chemicals and Materials, Nanjing Forestry
University, Nanjing 210037, China
| | - Aleksander Hejna
- Institute
of Materials Technology, Poznan University
of Technology, PL-61-138 Poznań, Poland
| | - Payam Zarrintaj
- School
of Chemical Engineering, Oklahoma State
University, 420 Engineering
North, Stillwater, Oklahoma 74078, United States
| | - Navid Rabiee
- Department
of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Justyna Kucinska-Lipka
- Department
of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, 80-233 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department
of Pharmaceutical Chemistry, Medical University
of Gdańsk, J.
Hallera 107, 80-416 Gdańsk, Poland
| | - Sidi A. Bencherif
- Chemical
Engineering Department, Northeastern University, Boston, Massachusetts 02115, United States
- Department
of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
- Harvard
John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
9
|
Simonini C, Natali P, Pirotti T, Nasillo V, Riva G, Chester J, Trenti T, Debbia D. SARS-CoV-2 infection is not associated with the emergence of monoclonal gammopathies. Int J Lab Hematol 2024; 46:451-456. [PMID: 38185475 DOI: 10.1111/ijlh.14225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Upon infection activated plasma cells produce large quantities of antibodies which can lead to the emergence of a monoclonal component (MC), detectable by serum protein electrophoresis (SPEP). This study aims to investigate any correlation between SARS-CoV-2 infection and MC development and, if identified, whether it persists during follow-up. METHODS SPEPs of 786 patients admitted to hospitals between March 01 2020 and March 31 2022 were evaluated. Positive (SARS-CoV-2+) and negative (SARS-CoV-2-) patients to nasopharyngeal swab for SARS-CoV-2 by RT-PCR were included. The persistence/new occurrence of MC was investigated for all patients during follow-up. Patient groups were compared by chi-square analysis. RESULTS MC was identified in 12% of all patients admitted to hospital, of which 28.7% were SARS-CoV-2+. The most common immunoglobulin isotype in both groups was IgG-k. There was no correlation between MC development and SARS-CoV-2 infection (p = 0.173). Furthermore, the risk of MC persistence in SARS-CoV-2-negative patients was revealed to be higher than in the SARS-CoV-2+ at follow-up (HR = 0.591, p = 0.05). CONCLUSIONS Our study suggests that the detection of MC during SARS-CoV-2 infection is most likely due to the hyperstimulation of the humoral immune system, as also occurs in other viral infections.
Collapse
Affiliation(s)
- Cecilia Simonini
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Italy
| | - Patrizia Natali
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| | - Tommaso Pirotti
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| | - Vincenzo Nasillo
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| | - Johanna Chester
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| | - Daria Debbia
- Department of Laboratory Medicine and Pathology, AUSL/AOU, Modena, Italy
| |
Collapse
|
10
|
Mollazadeh S, Mahmoudi M, Mohammadi M, Kamal Kheder R, Nasiri Mahallati H, Sarbaz Haghighi S, Masoumi E, Javanmardi Z, Esmaeili SA. Investigation of IL-6 serum level in COVID-19 patients with positive COVID-19 IgG/IgM antibody titers to check inflammation and disease progression. Cytokine 2024; 177:156564. [PMID: 38432065 DOI: 10.1016/j.cyto.2024.156564] [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: 02/01/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE The SARS-CoV-2, the cause of coronavirus disease 2019 (COVID-19), leads to severe pathogenicity and high mortality among different communities around the world. Therefore, it is important to understand the mechanisms of virus pathogenesis and the immune system's response to prevent the further spread of this virus. This study was aimed to evaluate the relationship between the serum level of interleukin 6 and positive IgG and IgM antibody levels in patients with COVID-19 to investigate inflammation and disease progression. METHODS & MATERIALS In this study, 10 ml of EDTA blood samples were taken from 414 COVID-19 patients. Then, the plasma was separated and the levels of IgM and IgG antibodies and interleukin 6 cytokine were evaluated by ELISA and chemiluminescence methods, respectively. All data were analyzed by SPSS 22 and GraphPad prism 9 software at the significance level of P < 0.05. RESULTS The results of this study showed that there was no significant difference in the expression of IgM and IgG antibodies between men and women. Also, a significant increase in the mean expression of IL-6 was observed only in the high concentration range (100-〉1000 pg/ml) in men compared to women (P < 0.001). In addition, in the female population, all three concentration ranges (negative, medium, and high) of IL-6 have the highest correlation with high titers (>10 U/ml) of IgM and IgG antibodies. While, in men, all three concentration ranges of IL-6 had the highest correlation with > 10 U/ml IgM antibody titers, but in the case of IgG, the highest correlation between different concentrations of IL-6 was observed with the negative or moderate titers of this antibody and there was an inverse relationship with the high titers of IgG (>10 U/ml). CONCLUSION As a result, the relationship between different serum levels of cytokine IL-6 with different titers of IgM and IgG antibodies was observed in both male and female populations. In general, it can be concluded that the correlation between different concentrations of IL-6 with different IgM titers was similar in both men and women, but in the case of different IgG titers, this correlation was higher in women than men.
Collapse
Affiliation(s)
- Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | | | | | - Elnaz Masoumi
- Hematology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Javanmardi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Teshnizi SH, Mirzazadeh S, Mashhadi N, Meri S, Kabelitz D, Kalantar K. Association study between killer immunoglobulin-like receptor polymorphisms and susceptibility to COVID-19 disease: a systematic review and meta-analysis. Immunol Res 2024; 72:175-184. [PMID: 37874432 DOI: 10.1007/s12026-023-09428-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a known virus that leads to a respiratory disease called coronavirus disease 19 (COVID-19). Natural killer (NK) cells, as members of innate immunity, possess crucial roles in restricting viral infections, including COVID-19. Their functions and development depend on receiving signals through various receptors, of which killer cell immunoglobulin-like receptors (KIRs) belong to the most effective ones. Different studies investigated the association between KIR gene content and susceptibility to COVID-19. Since previous studies have yielded contradictory results, we designed this meta-analysis study to draw comprehensive conclusions about COVID-19 risk and KIR gene association. According to PRISMA guidelines, a systematic search was performed in the electronic databases to find all studies investigating KIR gene contents in COVID-19 patients before March 2023. Any association between KIR genes and COVID-19 risk was determined by calculating pooled odds ratio (OR) and 95% confidence interval (CI). After applying the inclusion and exclusion criteria, 1673 COVID-19 patients and 1526 healthy controls from eight studies were included in this meta-analysis. As the main results, we observed a positive association between the 2DL3 (OR = 1.48, 95% CI = 1.17-1.88, P < 0.001) and susceptibility to COVID-19 and a negative association between the 2DP1 and the risk for COVID-19 (OR = 0.48, 95% CI = 0.23-0.99, P = 0.049). This meta-analysis demonstrated that KIR2DL3, as a member of iKIRs, might be associated with an increased risk of COVID-19 disease.
Collapse
Affiliation(s)
| | - Sara Mirzazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, PO Box: 71345-1798, Shiraz, Iran
| | - Niloofar Mashhadi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, PO Box: 71345-1798, Shiraz, Iran
| | - Seppo Meri
- Department of Bacteriology and Immunology and the Translational Immunology Research Program (TRIMM), The University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig, Holstein Campus Kiel, 24105, Kiel, Germany
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, PO Box: 71345-1798, Shiraz, Iran.
- Department of Bacteriology and Immunology and the Translational Immunology Research Program (TRIMM), The University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland.
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Wen H, Miao W, Liu B, Chen S, Zhang JS, Chen C, Quan MY. SPAUTIN-1 alleviates LPS-induced acute lung injury by inhibiting NF-κB pathway in neutrophils. Int Immunopharmacol 2024; 130:111741. [PMID: 38394887 DOI: 10.1016/j.intimp.2024.111741] [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: 12/22/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is an inflammatory condition characterized by acute damage to lung tissue. SPAUTIN-1, recognized as a small molecule drug targeting autophagy and USP10/13, has been reported for its potential to inhibit oxidative stress damage in various tissue injuries. However, the role and mechanism of SPAUTIN-1 in ALI remain unclear. This study aims to elucidate the protective effects of SPAUTIN-1 on ALI, with a particular focus on its role and mechanism in pulmonary inflammatory responses. METHODS Lipopolysaccharides (LPS) were employed to induce inflammation-mediated ALI. Bleomycin was used to induce non-inflammation-mediated ALI. The mechanism of SPAUTIN-1 action was identified through RNA-Sequencing and subsequently validated in mouse primary cells. Tert-butyl hydroperoxide (TBHP) was utilized to create an in vitro model of lung epithelial cell oxidative stress with MLE-12 cells. RESULTS SPAUTIN-1 significantly mitigated LPS-induced lung injury and inflammatory responses, attenuated necroptosis and apoptosis in lung epithelial cells, and inhibited autophagy in leukocytes and epithelial cells. However, SPAUTIN-1 exhibited no significant effect on bleomycin-induced lung injury. RNA-sequencing results demonstrated that SPAUTIN-1 significantly inhibited the NF-κB signaling pathway in leukocytes, a finding consistently confirmed by mouse primary cell assays. In vitro experiments further revealed that SPAUTIN-1 effectively mitigated oxidative stress injury in MLE-12 cells induced by TBHP. CONCLUSION SPAUTIN-1 alleviated LPS-induced inflammatory injury by inhibiting the NF-κB pathway in leukocytes and protected epithelial cells from oxidative damage, positioning it as a potential therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Hezhi Wen
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Wanqi Miao
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bin Liu
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shiyin Chen
- Wenzhou Medical University, Wenzhou 325000, China
| | - Jin-San Zhang
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Chengshui Chen
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Mei-Yu Quan
- Zhejiang Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
13
|
Dewi IP, Dachriyanus, Aldi Y, Ismail NH, Hefni D, Susanti M, Putra PP, Wahyuni FS. Comprehensive studies of the anti-inflammatory effect of tetraprenyltoluquinone, a quinone from Garcinia cowa Roxb. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117381. [PMID: 37967776 DOI: 10.1016/j.jep.2023.117381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Garcinia cowa Roxb. is called asam kandis in West Sumatra. This plant contains several quinone compounds, including tetraprenyltoluquinone (TPTQ). The bioactivity of this compound has been tested as an anticancer agent. However, reports regarding its anti-inflammatory effects are still limited, especially against coronavirus disease (Covid-19). AIM OF THE STUDY This study explores the anti-inflammatory effect of TPTQ in silico, in vitro, and in vivo. MATERIALS AND METHODS In silico testing used the Gnina application, opened via Google Colab. The TPTQ structure was docked with the nuclear factor kappa B (NF-ĸB) protein (PDB: 2RAM). In vitro testing began with testing the cytotoxicity of TPTQ against Raw 264.7 cells, using the 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) method. A phagocytic activity test was carried out using the neutral red uptake method, and interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) secretion tests were carried out using the enzyme-linked immunosorbent assay (ELISA) method. In vivo, tests were carried out on mice by determining cluster of differentiation 8+ (CD8+), natural killer cell (NK cell), and IL-6 parameters, using the ELISA method. RESULTS TPTQ has a lower binding energy than the native ligand and occupies the same active site as the native ligand. TPTQ decreased the phagocytosis index and secretion of IL-6 and TNF-α experimentally in vitro. TPTQ showed significant downregulation of CD8+ and slightly decreased NK cells and IL-6 secretion in vivo. CONCLUSION The potent inhibitory effect of TPTQ on the immune response suggests that TPTQ can be developed as an anti-inflammatory agent, especially in the treatment of Covid-19.
Collapse
Affiliation(s)
- Irene Puspa Dewi
- Faculty of Pharmacy, Universitas Andalas, Padang, 25163, Indonesia; Akademi Farmasi Prayoga, Padang, 25111, Indonesia
| | - Dachriyanus
- Faculty of Pharmacy, Universitas Andalas, Padang, 25163, Indonesia
| | - Yufri Aldi
- Faculty of Pharmacy, Universitas Andalas, Padang, 25163, Indonesia
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, UiTM Puncak Alam Campus, Selangor, Malaysia
| | - Dira Hefni
- Faculty of Pharmacy, Universitas Andalas, Padang, 25163, Indonesia
| | - Meri Susanti
- Faculty of Pharmacy, Universitas Andalas, Padang, 25163, Indonesia
| | | | | |
Collapse
|
14
|
Rieder AS, Wyse ATS. Regulation of Inflammation by IRAK-M Pathway Can Be Associated with nAchRalpha7 Activation and COVID-19. Mol Neurobiol 2024; 61:581-592. [PMID: 37640915 DOI: 10.1007/s12035-023-03567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023]
Abstract
In spite of the vaccine development and its importance, the SARS-CoV-2 pandemic is still impacting the world. It is known that the COVID-19 severity is related to the cytokine storm phenomenon, being inflammation a common disease feature. The nicotinic cholinergic system has been widely associated with COVID-19 since it plays a protective role in inflammation via nicotinic receptor alpha 7 (nAchRalpha7). In addition, SARS-CoV-2 spike protein (Spro) subunits can interact with nAchRalpha7. Moreover, Spro causes toll-like receptor (TLR) activation, leading to pro- and anti-inflammatory pathways. The increase and maturation of the IL-1 receptor-associated kinase (IRAK) family are mediated by activation of membrane receptors, such as TLRs. IRAK-M, a member of this family, is responsible for negatively regulating the activity of other active IRAKs. In addition, IRAK-M can regulate microglia phenotype by specific protein expression. Furthermore, there exists an antagonist influence of SARS-CoV-2 Spro and the cholinergic system action on the IRAK-M pathway and microglia phenotype. We discuss the overexpression and suppression of IRAK-M in inflammatory cell response to inflammation in SARS-CoV-2 infection when the cholinergic system is constantly activated via nAchRalpha7.
Collapse
Affiliation(s)
- Alessanda S Rieder
- Laboratory of Neuroprotection and Neurometabolic Diseases (Wyse's Lab), Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Neurometabolic Diseases (Wyse's Lab), Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre RS, 90035-003, Brazil.
| |
Collapse
|
15
|
Zhang T, Wang S, Liu Y, Qi X, Gao Y. Advances on adaptive immune responses affected by infectious bursal disease virus in chicken. Front Immunol 2024; 14:1330576. [PMID: 38268928 PMCID: PMC10806451 DOI: 10.3389/fimmu.2023.1330576] [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: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Infectious bursal disease (IBD) is an acute, highly infectious, and immunosuppressive disease caused by the infectious bursal disease virus (IBDV), which interferes with the immune system, causes hypoimmunity and seriously threatens the healthy development of the poultry industry. Adaptive immune response, an important defense line of host resistance to pathogen infection, is the host-specific immune response mainly mediated by T and B lymphocytes. As an important immunosuppressive pathogen in poultry, IBDV infection is closely related to the injury of the adaptive immune system. In this review, we focus on recent advances in adaptive immune response influenced by IBDV infection, especially the damage on immune organs, as well as the effect on humoral immune response and cellular immune response, hoping to provide a theoretical basis for further exploration of the molecular mechanism of immunosuppression induced by IBDV infection and the establishment of novel prevention and control measures for IBD.
Collapse
Affiliation(s)
- Tao Zhang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Suyan Wang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongzhen Liu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaole Qi
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yulong Gao
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
- National Poultry Laboratory Animal Resource Center, Harbin, China
| |
Collapse
|
16
|
Patra A, Bala A, Khan MR, Mukherjee AK. A Correlation Study to Comprehend the SAR-CoV-2 Viral Load, Antiviral Antibody Titer, and Severity of COVID-19 Symptoms Post-infection Amongst the Vaccinated Population in Kamrup District of As sam, Northeast India. Endocr Metab Immune Disord Drug Targets 2024; 24:1414-1421. [PMID: 38231052 DOI: 10.2174/0118715303281124231213110004] [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: 09/07/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND As per the recommendation of the United States Food and Drug Administration, more research is needed to determine the antibody titer against COVID-19 vaccination. OBJECTIVE The study aimed to understand the relationship between the antibody titer to the demographics, infection severity, and cycle threshold (CT) values of confirmed COVID-19 patients. METHODS Initially, we obtained consent from 185 populations and included sixty RT-PCRpositive COVID-19 patients from Kamrup District in the Northeast State of Assam, India. The vaccination status was recorded and tested for the level of serum immunoglobulin (IgG). The CT values, gender, and clinical symptoms-based scoring (CSBS) correlated with their IgG value. RESULTS Around 48% of participants gained an antibody titer more than the threshold value and showed CT values between 18-25. Moreover, the maximum distributed score above the average was found between the CT values 18-25. CONCLUSION The IgG titer value differs significantly amongst the vaccinated population, which may depend upon their genetic and demographic variability.
Collapse
Affiliation(s)
- Aparup Patra
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| | - Asis Bala
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| | - Mojibur R Khan
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| | - Ashis K Mukherjee
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| |
Collapse
|
17
|
Yan Y, Yang M, Jiao Y, Li L, Liu Z, Shi J, Shen Z, Peng G. Drug screening identified that handelin inhibits feline calicivirus infection by inhibiting HSP70 expression in vitro. J Gen Virol 2024; 105. [PMID: 38175184 DOI: 10.1099/jgv.0.001936] [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: 01/05/2024] Open
Abstract
Feline calicivirus (FCV) is considered one of the major pathogens of cats worldwide and causes upper respiratory tract disease in all cats. In some cats, infection is by a highly virulent strain of FCV (vs.-FCV), which can cause severe and fatal systemic disease symptoms. At present, few antiviral drugs are approved for clinical treatment against FCV. Therefore, there is an imminent need for effective FCV antiviral agents. Here, we used observed a cytopathic effect (CPE) assay to screen 1746 traditional Chinese medicine monomer compounds and found one that can effectively inhibit FCV replication, namely, handelin, with an effective concentration (EC50) value of approximately 2.5 µM. Further study showed that handelin inhibits FCV replication via interference with heat shock protein 70 (HSP70), which is a crucial host factor and plays a positive role in regulating viral replication. Moreover, handelin and HSP70 inhibitors have broad-spectrum antiviral activity. These findings indicate that handelin is a potential candidate for the treatment of FCV infection and that HSP70 may be an important drug target.
Collapse
Affiliation(s)
- Yuanyuan Yan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Mengfang Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Yuzhou Jiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Lisha Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Zirui Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Jiale Shi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Zhou Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| |
Collapse
|
18
|
Wang J, Wu Y, Lin R, Zhang Y, Li L. TRAM deletion attenuates monocyte exhaustion and alleviates sepsis severity. Front Immunol 2023; 14:1297329. [PMID: 38162637 PMCID: PMC10756061 DOI: 10.3389/fimmu.2023.1297329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Monocyte exhaustion characterized by immune-suppressive features can develop during sepsis and contribute to adverse patient outcomes. However, molecular mechanisms responsible for the establishment of immune-suppressive monocytes with reduced expression of immune-enhancing mediators such as CD86 during sepsis are not well understood. In this study, we identified that the TLR4 intracellular adaptor TRAM plays a key role in mediating the sustained reduction of CD86 expression on exhausted monocytes and generating an immune-suppressive monocyte state. TRAM contributes to the prolonged suppression of CD86 through inducing TAX1BP1 as well as SARM1, collectively inhibiting Akt and NFκB. TRAM deficient mice are protected from cecal slurry-induced experimental sepsis and retain immune-competent monocytes with CD86 expression. Our data reveal a key molecular circuitry responsible for monocyte exhaustion and provide a viable target for rejuvenating functional monocytes and treating sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
19
|
Park S, Choi BH, Jee YS. Effects of plank exercise on respiratory capacity, physical fitness, and immunocytes in older adults. J Exerc Rehabil 2023; 19:332-338. [PMID: 38188128 PMCID: PMC10766451 DOI: 10.12965/jer.2346536.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024] Open
Abstract
Plank is a readily accessible form of exercise that can benefit individuals of various fitness levels. However, its effectiveness in older individuals has not been conclusively established. Specifically, its impact on respiratory function, physical fitness, and immunocytes in them has not been thoroughly verified. The study encompassed participants with an average age of 64.33±1.98 years. All participants were randomly assigned to either the control group (COG) or the plank exercise group (PXG). The plank exercise was conducted 3 days a week for 12 weeks. While the values in the COG deteriorated, the PXG showed significant improvements in several parameters. Forced expiratory volume in one second and peak expiratory flow increased by approximately 27% and 16%, respectively, in the PXG, demonstrating significant differences (P<0.001) between the two groups. Additionally, VO2max, grip strength, and sit-ups increased by about 12%, 18%, and 42% in the PXG. Notably, innate immunocytes, such as NK cells, increased by approximately 30% in the PXG. For adaptive immunocytes, including CD3+ T cells, CD4+ T cells, and CD8+ T cells, there were notable increases of around 18%, 19%, and 28%, respectively, in the PXG. These findings underline significant differences (P<0.001) between the two groups. This study provides confirmation that engaging in plank exercise can enhance the function of immunocytes, while also improving respiratory capacity and physical fitness in older adults.
Collapse
Affiliation(s)
- Sihwa Park
- Research Institute of Sports and Industry Science, Hanseo University, Seosan,
Korea
| | - Byung-Hwa Choi
- Department of Physical Education, Graduate School of Hanseo University, Seosan,
Korea
| | - Yong-Seok Jee
- Research Institute of Sports and Industry Science, Hanseo University, Seosan,
Korea
- Department of Physical Education, Graduate School of Hanseo University, Seosan,
Korea
| |
Collapse
|
20
|
Song Y, Yao L, Li S, Zhou J. Psoriasis comorbidity management in the COVID era: a pressing challenge. Front Microbiol 2023; 14:1294056. [PMID: 38029150 PMCID: PMC10667470 DOI: 10.3389/fmicb.2023.1294056] [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: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The global COVID-19 pandemic has presented a significant, ongoing challenge since its emergence in late 2019. Today, the Omicron strain, which is less lethal but more contagious than the original outbreak strain, continues to pose substantial health risks. In this background, the management of psoriatic comorbidities has become even more complex, particularly for patients with underlying inflammatory, metabolic, or cardiovascular diseases. This review aims to summarize current research on comorbid COVID-19 and psoriasis, and provide insights into the development of evidence-based management strategies. By providing appropriate patient instruction, implementing protective measures, and re-evaluating medication prescriptions based on each patient's unique situation, healthcare professionals can effectively address the challenges faced by patients with comorbid psoriasis in the COVID-19 era.
Collapse
Affiliation(s)
| | | | | | - Junfeng Zhou
- Department of Dermatology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Yang X, Wang J, Liu W. Molecular markers of type II alveolar epithelial cells in acute lung injury by bioinformatics analysis. Sci Rep 2023; 13:17797. [PMID: 37853056 PMCID: PMC10584938 DOI: 10.1038/s41598-023-45129-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023] Open
Abstract
In this study, we aimed to identify molecular markers associated with type II alveolar epithelial cell injury in acute lung injury (ALI) models using bioinformatics methods. The objective was to provide new insights for the diagnosis and treatment of ALI/ARDS. We downloaded RNA SEQ datasets (GSE109913, GSE179418, and GSE119123) from the Gene Expression Omnibus (GEO) and used R language package to screen differentially expressed genes (DEGs). DEGs were annotated using Gene Ontology (GO), and their pathways were analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG). DEGs were imported into the STRING database and analyzed using Cytoscape software to determine the protein network of DEGs and calculate the top 10 nodes for the hub genes. Finally, potential therapeutic drugs for the hub genes were predicted using the DGIdb database. We identified 78 DEGs, including 70 up-regulated genes and 8 down-regulated genes. GO analysis revealed that the DEGs were mainly involved in biological processes such as granulocyte migration, response to bacterial-derived molecules, and cytokine-mediated signaling pathways. Additionally, they had cytokine activity, chemokine activity, and receptor ligand activity, and functioned in related receptor binding, CXCR chemokine receptor binding, G protein-coupled receptor binding, and other molecular functions. KEGG analysis indicated that the DEGs were mainly involved in TNF signaling pathway, IL-17 signaling pathway, NF-κB signal pathway, chemokine signal pathway, cytokine-cytokine receptor interaction signal pathway, and others. We identified eight hub genes, including IRF7, IFIT1, IFIT3, PSMB8, PSMB9, BST2, OASL2, and ZBP1, which were all up-regulated genes. We identified several hub genes of type II alveolar epithelial cells in ALI mouse models using bioinformatics analysis. These results provide new targets for understanding and treating of ALI.
Collapse
Affiliation(s)
- Xiaoting Yang
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Jing Wang
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Wei Liu
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
22
|
Rutkowska E, Kwiecień I, Pietruszka-Wałęka E, Więsik-Szewczyk E, Rzepecki P, Jahnz-Różyk K. Analysis of Leukocyte Subpopulations by Flow Cytometry during Hospitalization Depending on the Severity of COVID-19 Course. Biomedicines 2023; 11:2728. [PMID: 37893102 PMCID: PMC10604221 DOI: 10.3390/biomedicines11102728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The mechanisms underlying the immune response to coronavirus disease 2019 (COVID-19) and the recovery process have not been fully elucidated. The aim of the study was to analyze leukocyte subpopulations in patients at significant time points (at diagnosis, and 3 and 6 months after infection) selected according to the analysis of changes in the lungs by the CT classification system, considering the severity of the disease. The study groups consisted of severe and non-severe COVID-19 patients. There was a significant decrease in CD8+ T cells, NK and eosinophils, with an increasing percentage of neutrophils during hospitalization. We noticed lower levels of CD4 and CD8 T lymphocytes, eosinophils, basophils, and CD16+ monocytes and elevated neutrophil levels in severe patients relative to non-severe patients. Three months after infection, we observed higher levels of basophils, and after 6 months, higher CD4/CD8 ratios and T cell levels in the severe compared to non-severe group. Non-severe patients showed significant changes in the leukocyte populations studied at time of hospitalization and both within 3 months and 6 months of onset. The CT CSS classification with parameters of the flow cytometry method used for COVID-19 patients determined changes that proved useful in the initial evaluation of patients.
Collapse
Affiliation(s)
- Elżbieta Rutkowska
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Iwona Kwiecień
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Ewa Pietruszka-Wałęka
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Piotr Rzepecki
- Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| |
Collapse
|
23
|
Li H, Wang X, Wang Y, Li Y, Chen Y, Wong YT, He J, He ML. Secreted LRPAP1 binds and triggers IFNAR1 degradation to facilitate virus evasion from cellular innate immunity. Signal Transduct Target Ther 2023; 8:374. [PMID: 37743411 PMCID: PMC10518340 DOI: 10.1038/s41392-023-01630-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/04/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
The crucial role of interferon (IFN) signaling is well known in the restriction or eradication of pathogen invasion. Viruses take a variety of ways to antagonize host defense through eliminating IFN-signaling intracellularly for decades. However, the way by viruses target IFN-signaling extracellularly has not been discovered. Infection by both coronavirus SARS-CoV-2 and enterovirus 71 (EV71 or EV-A71) can cause severe diseases such as neurological disorders and even death in children.1-3 Here, we show evidence that the protease of SARS-CoV-2 (3CLpro) and EV71 (2Apro) upregulates the expression and secretion of LDL-receptor-related protein-associated protein 1 (LRPAP1). As a ligand, the N-terminus of secreted LRPAP1 binds with the extracellular domain of IFNAR1 that triggers the receptor ubiquitination and degradation and promotes virus infection both in vitro, ex vivo in the mouse brain, and in vivo in newborn mice. A small peptide from the N-terminus of LRPAP1 effectively binds and causes IFNAR1 degradation that enhances both DNA and RNA viral infections, including herpesvirus HSV-1, hepatitis B virus (HBV), EV71, and beta-coronavirus HCoV-OC43; whereas α2M, a LRPAP1 inhibitor, arrests virus infections by stabilizing IFNAR1. Our study demonstrates a new mechanism used by viruses for evading host cell immunity, supporting a strategy for developing pan-antiviral drugs.
Collapse
Affiliation(s)
- Huangcan Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- CityU Shenzhen Research Institute, Nanshan, Shenzhen, China
| | - Xiong Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yiran Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yichen Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yin-Ting Wong
- Department of Neurosciences, City University of Hong Kong, Hong Kong, China
| | - Jufang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Department of Neurosciences, City University of Hong Kong, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
- CityU Shenzhen Research Institute, Nanshan, Shenzhen, China.
| |
Collapse
|
24
|
Isazadeh A, Heris JA, Shahabi P, Mohammadinasab R, Shomali N, Nasiri H, Valedkarimi Z, Khosroshahi AJ, Hajazimian S, Akbari M, Sadeghvand S. Pattern-recognition receptors (PRRs) in SARS-CoV-2. Life Sci 2023; 329:121940. [PMID: 37451397 DOI: 10.1016/j.lfs.2023.121940] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Pattern recognition receptors (PRRs) are specific sensors that directly recognize various molecules derived from viral or bacterial pathogens, senescent cells, damaged cells, and apoptotic cells. These sensors act as a bridge between nonspecific and specific immunity in humans. PRRs in human innate immunity were classified into six types: toll-like receptors (TLR), C-type lectin receptors (CLRs), nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs), absent in melanoma 2 (AIM2)-like receptors (ALRs), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), and cyclic GMP-AMP (cGAMP) synthase (cGAS). Numerous types of PRRs are responsible for recognizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which is immensely effective in prompting interferon responses. Detection of SARS-CoV-2 infection by PRRs causes the initiation of an intracellular signaling cascade and subsequently the activation of various transcription factors that stimulate the production of cytokines, chemokines, and other immune-related factors. Therefore, it seems that PRRs are a promising potential therapeutic approach for combating SARS-CoV-2 infection and other microbial infections. In this review, we have introduced the current knowledge of various PRRs and related signaling pathways in response to SARS-CoV-2.
Collapse
Affiliation(s)
- Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Valedkarimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Shahram Sadeghvand
- Department of Pediatrics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
25
|
Li D, Quan Z, Ni J, Li H, Qing H. The many faces of the zinc finger protein 335 in brain development and immune system. Biomed Pharmacother 2023; 165:115257. [PMID: 37541176 DOI: 10.1016/j.biopha.2023.115257] [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: 06/02/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
Zinc finger protein 335 (ZNF335) plays a crucial role in the methylation and, consequently, regulates the expression of a specific set of genes. Variants of the ZNF335 gene have been identified as risk factors for microcephaly in a variety of populations worldwide. Meanwhile, ZNF335 has also been identified as an essential regulator of T-cell development. However, an in-depth understanding of the role of ZNF335 in brain development and T cell maturation is still lacking. In this review, we summarize current knowledge of the molecular mechanisms underlying the involvement of ZNF335 in neuronal and T cell development across a wide range of pre-clinical, post-mortem, ex vivo, in vivo, and clinical studies. We also review the current limitations regarding the study of the pathophysiological functions of ZNF335. Finally, we hypothesize a potential role for ZNF335 in brain disorders and discuss the rationale of targeting ZNF335 as a therapeutic strategy for preventing brain disorders.
Collapse
Affiliation(s)
- Danyang Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
26
|
Jing H, Chen X, Wang D. Identification of biomarkers associated with diagnosis of acute lung injury based on bioinformatics and machine learning. Medicine (Baltimore) 2023; 102:e34840. [PMID: 37603512 PMCID: PMC10443773 DOI: 10.1097/md.0000000000034840] [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: 02/24/2023] [Accepted: 07/28/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is an acute inflammatory disease characterized by excess production of inflammatory factors in lung tissue and has a high mortality. This research was designed for the identification of novel diagnostic biomarkers for ALI and analyzing the possible association between critical genes and infiltrated immune cells. METHODS The study used 2 datasets (GSE2411 and GSE18341) to identify differentially expressed genes (DEGs) between 2 groups. Then we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses to identify the functions of these DEGs. The study also used SVM-recursive feature elimination analysis and least absolute shrinkage and selection operator regression model to screen possible markers. The study further analyzed immune cell infiltration via CIBERSORT. Gene Set Enrichment Analysis was used to explore the molecular mechanism of the critical genes. RESULTS DEGs were identified between 2 groups. In total, 690 DEGs were obtained: 527 genes were upregulated and 163 genes were downregulated. We identified PDZK1IP1, CCKAR, and CXCL2 as critical genes. And we then found that these critical genes correlated with Mast Cells, Neutrophil Cells, M1 Macrophage, dendritic cell Actived, Eosinophil Cells, B Cells Naive, Mast Cells, and dendritic cell Immature. Furthermore, we investigated the specific signaling pathways involved in key genes and derived some potential molecular mechanisms by which key genes affect disease progression by use of Gene Set Enrichment Analysis. Moreover, we predict transcription factors. Also, we obtained critical gene-related microRNAs through the targetscan database, and visualized the microRNA network of the genes. CONCLUSION Our findings might provide some novel clue for the exploration of novel markers for ALI diagnosis. The critical genes and their associations with immune infiltration may offer new insight into understanding ALI developments.
Collapse
Affiliation(s)
- Hekun Jing
- Department of Respiratory and Critical Care of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaorui Chen
- Department of Respiratory and Critical Care of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daoxin Wang
- Department of Respiratory and Critical Care of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Qu M, Zhang H, Cheng P, Wubshet AK, Yin X, Wang X, Sun Y. Histone deacetylase 6's function in viral infection, innate immunity, and disease: latest advances. Front Immunol 2023; 14:1216548. [PMID: 37638049 PMCID: PMC10450946 DOI: 10.3389/fimmu.2023.1216548] [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: 05/04/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
In the family of histone-deacetylases, histone deacetylase 6 (HDAC6) stands out. The cytoplasmic class IIb histone deacetylase (HDAC) family is essential for many cellular functions. It plays a crucial and debatable regulatory role in innate antiviral immunity. This review summarises the current state of our understanding of HDAC6's structure and function in light of the three mechanisms by which it controls DNA and RNA virus infection: cytoskeleton regulation, host innate immune response, and autophagy degradation of host or viral proteins. In addition, we summed up how HDAC6 inhibitors are used to treat a wide range of diseases, and how its upstream signaling plays a role in the antiviral mechanism. Together, the findings of this review highlight HDAC6's importance as a new therapeutic target in antiviral immunity, innate immune response, and some diseases, all of which offer promising new avenues for the development of drugs targeting the immune response.
Collapse
Affiliation(s)
- Min Qu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huijun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengyuan Cheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ashenafi Kiros Wubshet
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Basic and Diagnostic Sciences, College of Veterinary Science, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
28
|
Apostolo D, Ferreira LL, Di Tizio A, Ruaro B, Patrucco F, Bellan M. A Review: The Potential Involvement of Growth Arrest-Specific 6 and Its Receptors in the Pathogenesis of Lung Damage and in Coronavirus Disease 2019. Microorganisms 2023; 11:2038. [PMID: 37630598 PMCID: PMC10459962 DOI: 10.3390/microorganisms11082038] [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: 06/21/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
The tyrosine kinase receptors of the TAM family-Tyro3, Axl and Mer-and their main ligand Gas6 (growth arrest-specific 6) have been implicated in several human diseases, having a particularly important role in the regulation of innate immunity and inflammatory response. The Gas6/TAM system is involved in the recognition of apoptotic debris by immune cells and this mechanism has been exploited by viruses for cell entry and infection. Coronavirus disease 2019 (COVID-19) is a multi-systemic disease, but the lungs are particularly affected during the acute phase and some patients may suffer persistent lung damage. Among the manifestations of the disease, fibrotic abnormalities have been observed among the survivors of COVID-19. The mechanisms of COVID-related fibrosis remain elusive, even though some parallels may be drawn with other fibrotic diseases, such as idiopathic pulmonary fibrosis. Due to the still limited number of scientific studies addressing this question, in this review we aimed to integrate the current knowledge of the Gas6/TAM axis with the pathophysiological mechanisms underlying COVID-19, with emphasis on the development of a fibrotic phenotype.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
| | - Luciana L. Ferreira
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
| | - Alice Di Tizio
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
- Respiratory Diseases Unit, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Barbara Ruaro
- Pulmonology Department, University of Trieste, 34128 Trieste, Italy;
| | - Filippo Patrucco
- Respiratory Diseases Unit, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
- Division of Internal Medicine, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| |
Collapse
|
29
|
Liang Q, Wang L, Xu J, Lin A, Wu Y, Tao Q, Zhang B, Min H, Song S, Gao Q. A burns and COVID-19 shared stress responding gene network deciphers CD1C-CD141- DCs as the key cellular components in septic prognosis. Cell Death Discov 2023; 9:258. [PMID: 37488118 PMCID: PMC10366195 DOI: 10.1038/s41420-023-01518-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
Differential body responses to various stresses, infectious or noninfectious, govern clinical outcomes ranging from asymptoma to death. However, the common molecular and cellular nature of the stress responsome across different stimuli is not described. In this study, we compared the expression behaviors between burns and COVID-19 infection by choosing the transcriptome of peripheral blood from related patients as the analytic target since the blood cells reflect the systemic landscape of immune status. To this end, we identified an immune co-stimulator (CD86)-centered network, named stress-response core (SRC), which was robustly co-expressed in burns and COVID-19. The enhancement of SRC genes (SRCs) expression indicated favorable prognosis and less severity in both conditions. An independent whole blood single-cell RNA sequencing of COVID-19 patients demonstrated that the monocyte-dendritic cell (Mono-DC) wing was the major cellular source of SRC, among which the higher expression of the SRCs in the monocyte was associated with the asymptomatic COVID-19 patients, while the quantity-restricted and function-defected CD1C-CD141-DCs were recognized as the key signature which linked to bad consequences. Specifically, the proportion of the CD1C-CD141-DCs and their SRCs expression were step-wise reduced along with worse clinic conditions while the subcluster of CD1C-CD141-DCs from the critical COVID-19 patients was characterized of IFN signaling quiescence, high mitochondrial metabolism and immune-communication inactivation. Thus, our study identified an expression-synchronized and function-focused gene network in Mono-DC population whose expression status was prognosis-related and might serve as a new target of diagnosis and therapy.
Collapse
Affiliation(s)
- Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Lei Wang
- Department of Clinical Laboratory, Jiangsu Provincial Hospital of Integrated Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Jing Xu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Anqi Lin
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Yongzheng Wu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Bin Zhang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
- Central Laboratory, Nanjing Chest Hospital, Nanjing Medical University, Nanjing, 210028, China
| | - Haiyan Min
- Central Laboratory, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China.
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China.
| |
Collapse
|
30
|
Xu J, Li XX, Yuan N, Li C, Yang JG, Cheng LM, Lu ZX, Hou HY, Zhang B, Hu H, Qian Y, Liu XX, Li GC, Wang YD, Chu M, Dong CR, Liu F, Ge QG, Yang YJ. T cell receptor β repertoires in patients with COVID-19 reveal disease severity signatures. Front Immunol 2023; 14:1190844. [PMID: 37475855 PMCID: PMC10355153 DOI: 10.3389/fimmu.2023.1190844] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/06/2023] [Indexed: 07/22/2023] Open
Abstract
Background The immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are crucial in maintaining a delicate balance between protective effects and harmful pathological reactions that drive the progression of coronavirus disease 2019 (COVID-19). T cells play a significant role in adaptive antiviral immune responses, making it valuable to investigate the heterogeneity and diversity of SARS-CoV-2-specific T cell responses in COVID-19 patients with varying disease severity. Methods In this study, we employed high-throughput T cell receptor (TCR) β repertoire sequencing to analyze TCR profiles in the peripheral blood of 192 patients with COVID-19, including those with moderate, severe, or critical symptoms, and compared them with 81 healthy controls. We specifically focused on SARS-CoV-2-associated TCR clonotypes. Results We observed a decrease in the diversity of TCR clonotypes in COVID-19 patients compared to healthy controls. However, the overall abundance of dominant clones increased with disease severity. Additionally, we identified significant differences in the genomic rearrangement of variable (V), joining (J), and VJ pairings between the patient groups. Furthermore, the SARS-CoV-2-associated TCRs we identified enabled accurate differentiation between COVID-19 patients and healthy controls (AUC > 0.98) and distinguished those with moderate symptoms from those with more severe forms of the disease (AUC > 0.8). These findings suggest that TCR repertoires can serve as informative biomarkers for monitoring COVID-19 progression. Conclusions Our study provides valuable insights into TCR repertoire signatures that can be utilized to assess host immunity to COVID-19. These findings have important implications for the use of TCR β repertoires in monitoring disease development and indicating disease severity.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital & National Center for Cardiovascular Diseases, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiao-xiao Li
- Department of Pharmacy and Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Na Yuan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chao Li
- Department of Pharmacy and Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Jin-gang Yang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital & National Center for Cardiovascular Diseases, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li-ming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhong-xin Lu
- Department of Medical Laboratory, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-yan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Zhang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Hu
- Department of Medical Laboratory, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Qian
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin-xuan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guo-chao Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
| | - Yue-dan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Chao-ran Dong
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University of Security Sciences, Riyadh, Saudi Arabia
| | - Qing-gang Ge
- Department of Pharmacy and Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Yue-jin Yang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital & National Center for Cardiovascular Diseases, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Jensen GS, Yu L, Iloba I, Cruickshank D, Matos JR, Newman RA. Differential Activities of the Botanical Extract PBI-05204 and Oleandrin on Innate Immune Functions under Viral Challenge Versus Inflammatory Culture Conditions. Molecules 2023; 28:4799. [PMID: 37375354 DOI: 10.3390/molecules28124799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The Nerium oleander extract PBI 05204 (PBI) and its cardiac glycoside constituent oleandrin have direct anti-viral properties. Their effect on the immune system, however, is largely unknown. We used an in vitro model of human peripheral blood mononuclear cells to document effects under three different culture conditions: normal, challenged with the viral mimetic polyinosinic:polycytidylic acid Poly I:C, and inflamed by lipopolysaccharide (LPS). Cells were evaluated for immune activation marks CD69, CD25, and CD107a, and culture supernatants were tested for cytokines. Both PBI and oleandrin directly activated Natural Killer (NK) cells and monocytes and triggered increased production of cytokines. Under viral mimetic challenge, PBI and oleandrin enhanced the Poly I:C-mediated immune activation of monocytes and NK cells and enhanced production of IFN-γ. Under inflammatory conditions, many cytokines were controlled at similar levels as in cultures treated with PBI and oleandrin without inflammation. PBI triggered higher levels of some cytokines than oleandrin. Both products increased T cell cytotoxic attack on malignant target cells, strongest by PBI. The results show that PBI and oleandrin directly activate innate immune cells, enhance anti-viral immune responses through NK cell activation and IFN-γ levels, and modulate immune responses under inflamed conditions. The potential clinical impact of these activities is discussed.
Collapse
Affiliation(s)
| | - Liu Yu
- NIS Labs, 807 St. George St., Port Dover, ON N0A 1N0, Canada
| | - Ifeanyi Iloba
- NIS Labs, 1437 Esplanade, Klamath Falls, OR 97601, USA
| | | | - Jose R Matos
- Phoenix Biotechnology, 8626 Tesoro Drive, Suite 801, San Antonio, TX 78217, USA
| | - Robert A Newman
- Phoenix Biotechnology, 8626 Tesoro Drive, Suite 801, San Antonio, TX 78217, USA
| |
Collapse
|
32
|
Russo C, Valle MS, Malaguarnera L, Romano IR, Malaguarnera L. Comparison of Vitamin D and Resveratrol Performances in COVID-19. Nutrients 2023; 15:nu15112639. [PMID: 37299603 DOI: 10.3390/nu15112639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Over the last few years, we have experienced the infection generated by severe respiratory syndrome coronavirus 2 (SARS-CoV-2) often resulting in an exaggerated immune reaction and systemic inflammation. The preferred treatments against SARS-CoV-2 were those that mitigated immunological/inflammatory dysfunction. A variety of observational epidemiological studies have reported that vitamin D deficiency is often a crucial factor in many inflammatory diseases and autoimmune diseases, as well as the susceptibility to contract infectious diseases, including acute respiratory infections. Similarly, resveratrol regulates immunity, modifying the gene expression and the release of proinflammatory cytokines in the immune cells. Therefore, it plays an immunomodulatory role that can be beneficial in the prevention and development of non-communicable diseases associated with inflammation. Since both vitamin D and resveratrol also act as immunomodulators in inflammatory pathologies, many studies have paid particular attention to an integrated treatment of either vitamin D or resveratrol in the immune reaction against SARS-CoV-2 infections. This article offers a critical evaluation of published clinical trials that have examined the use of vitamin D or resveratrol as adjuncts in COVID-19 management. Furthermore, we aimed to compare the anti-inflammatory and antioxidant properties linked to the modulation of the immune system, along with antiviral properties of both vitamin D and resveratrol.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Luisa Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Ivana Roberta Romano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
33
|
Chen B, Xue Y, Jing H, Wang X, Zhu P, Hao W, Li M, Gao Y. Effectiveness of Chinese medicine formula Huashibaidu granule on mild COVID-19 patients: A prospective, non-randomized, controlled trial. Integr Med Res 2023; 12:100950. [PMID: 37192979 PMCID: PMC10121152 DOI: 10.1016/j.imr.2023.100950] [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: 09/14/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/18/2023] Open
Abstract
Background The effectiveness and safety of Huashibaidu granule (HSBD) in treating mild Corona Virus Disease 2019 (COVID-19) patients infected with SARS-CoV-2 remain to be identified. We aimed to evaluate the effectiveness of HSBD in mild COVID-19 patients. Methods A prospective, non-randomized, controlled study in mild COVID-19 patients was conducted in Shanghai, from April 8 to May 6, 2022. The enrolled patients were diagnosed as mild COVID-19. Finally, 360 patients received HSBD, and 368 patients received TCM placebo (administered orally 20 g twice daily for 7 days). The primary endpoints were the negative conversion rate of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and the negative conversion time. Secondary endpoints included the hospitalized days and the improvement in the clinical condition. Results The negative conversion rate of SARS-CoV-2 at 7 days posttreatment in the HSBD group was higher than that in the control group (95.28% vs. 82.61%, P < 0.001). The median negative conversion time in the HSBD group was markedly decreased by 2 days compared with the control group (3 [3-6] vs. 5 [4-7], P < 0.001). In addition, the median hospitalized day was shortened in the HSBD group by 1 day compared with the control group (6 [4-7] vs. 7 [5-9], P < 0.001). The clinical improvement rate (275/360 [76.39%]) in the HSBD group within 7 days was significantly higher than that (203/368 [55.16%]) in the control group (P < 0.001). The improvement of symptom scores in the HSBD group was higher than that in the control group (2 [1-4] vs. 1 [1-2], P < 0.001). No severe adverse events occurred. Conclusions Our study suggested that HSBD effectively increased the negative conversion rate of SARS-CoV-2 and shortened the negative conversion time and hospitalized days in mild COVID-19 patients. Clinical trial registration Chinese Clinical Trial Registry, ChiCTR2200058668.
Collapse
Affiliation(s)
- Bowu Chen
- Department of Hepatology, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Xue
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Jing
- Naval Medical Center, Shanghai, China
| | - Xiaodong Wang
- Nursing Department, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peimin Zhu
- Department of Gastroenterology, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Hao
- Department of Gastroenterology, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Li
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueqiu Gao
- Department of Hepatology, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| |
Collapse
|
34
|
Meo C, Palma G, Bruzzese F, Budillon A, Napoli C, de Nigris F. Spontaneous cancer remission after COVID-19: insights from the pandemic and their relevance for cancer treatment. J Transl Med 2023; 21:273. [PMID: 37085802 PMCID: PMC10119533 DOI: 10.1186/s12967-023-04110-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Early in the COVID-19 pandemic, it emerged that the risk of severe outcomes was greater in patients with co-morbidities, including cancer. The huge effort undertaken to fight the pandemic, affects the management of cancer care, influencing their outcome. Despite the high fatality rate of COVID-19 disease in cancer patients, rare cases of temporary or prolonged clinical remission from cancers after SARS-CoV-2 infection have been reported. We have reviewed sixteen case reports of COVID-19 disease with spontaneous cancer reduction of progression. Fourteen cases of remission following viral infections and two after anti-SARS-CoV-2 vaccination. The immune response to COVID-19, may be implicated in both tumor regression, and progression. Specifically, we discuss potential mechanisms which include oncolytic and priming hypotheses, that may have contributed to the cancer regression in these cases and could be useful for future options in cancer treatment.
Collapse
Affiliation(s)
- Concetta Meo
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via De Crecchio 7, 80138, Naples, Italy
| | - Giuseppe Palma
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Francesca Bruzzese
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate - National Institute of Cancer - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistic Units, Division of Clinical Immunology and Immunohematology, Transfusion Medicine, and Transplant Immunology (SIMT), Azienda Universitaria Policlinico (AOU), 80138, Naples, Italy
- Advanced Medical and Surgical Science (DAMSS), School of Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Filomena de Nigris
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
35
|
Gonçalves‐Pereira MH, Santiago L, Ravetti CG, Vassallo PF, de Andrade MVM, Vieira MS, de Fátima Souza de Oliveira F, Carobin NV, Li G, de Paula Sabino A, Nobre V, da Costa Santiago H. Dysfunctional phenotype of systemic and pulmonary regulatory T cells associate with lethal COVID-19 cases. Immunology 2023; 168:684-696. [PMID: 36349514 PMCID: PMC9877711 DOI: 10.1111/imm.13603] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
Severe cases of COVID-19 present hyperinflammatory condition that can be fatal. Little is known about the role of regulatory responses in SARS-CoV-2 infection. In this study, we evaluated the phenotype of regulatory T cells in the blood (peripheral blood mononuclear cell) and the lungs (broncho-alveolar) of adult patients with severe COVID-19 under invasive mechanical ventilation. Our results show important dynamic variation on Treg cells phenotype during COVID-19 with changes in number and functional parameters from the day of intubation (Day 1 of intensive care unit admission) to Day 7. We observed that compared with surviving patients, non-survivors presented lower numbers of Treg cells in the blood. In addition, lung Tregs of non-survivors also displayed higher PD1 and lower FOXP3 expressions suggesting dysfunctional phenotype. Further signs of Treg dysregulation were observed in non-survivors such as limited production of IL-10 in the lungs and higher production of IL-17A in the blood and in the lungs, which were associated with increased PD1 expression. These findings were also associated with lower pulmonary levels of Treg-stimulating factors like TNF and IL-2. Tregs in the blood and lungs are profoundly dysfunctional in non-surviving COVID-19 patients.
Collapse
Affiliation(s)
- Marcela Helena Gonçalves‐Pereira
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Luciana Santiago
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Hospital das ClínicasUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Núcleo Interdisciplinar de Investigação em Medicina IntensivaDepartamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Cecilia Gómez Ravetti
- Núcleo Interdisciplinar de Investigação em Medicina IntensivaDepartamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Faculdade de MedicinaUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Paula Frizera Vassallo
- Núcleo Interdisciplinar de Investigação em Medicina IntensivaDepartamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Faculdade de MedicinaUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Marcus Vinicius Melo de Andrade
- Núcleo Interdisciplinar de Investigação em Medicina IntensivaDepartamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Faculdade de MedicinaUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Mariana Sousa Vieira
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | | | - Natália Virtude Carobin
- Departamento de Análises Clínicas e ToxicológicasFaculdade de Farmácia, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Guangzhao Li
- Department of MicrobiologyImmunology and Tropical Medicine, The George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Adriano de Paula Sabino
- Departamento de Análises Clínicas e ToxicológicasFaculdade de Farmácia, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Vandack Nobre
- Núcleo Interdisciplinar de Investigação em Medicina IntensivaDepartamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Faculdade de MedicinaUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| |
Collapse
|
36
|
Thakur A. Shedding Lights on the Extracellular Vesicles as Functional Mediator and Therapeutic Decoy for COVID-19. Life (Basel) 2023; 13:life13030840. [PMID: 36983995 PMCID: PMC10052528 DOI: 10.3390/life13030840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
COVID-19 is an infectious disease caused by the novel coronavirus (SARS-CoV-2) that first appeared in late 2019 and has since spread across the world. It is characterized by symptoms such as fever, cough, and shortness of breath and can lead to death in severe cases. To help contain the virus, measures such as social distancing, handwashing, and other public health measures have been implemented. Vaccine and drug candidates, such as those developed by Pfizer/BioNTech, AstraZeneca, Moderna, Novavax, and Johnson & Johnson, have been developed and are being distributed worldwide. Clinical trials for drug treatments such as remdesivir, dexamethasone, and monoclonal antibodies are underway and have shown promising results. Recently, exosomes have gained attention as a possible mediator of the COVID-19 infection. Exosomes, small vesicles with a size of around 30-200 nm, released from cells, contain viral particles and other molecules that can activate the immune system and/or facilitate viral entry into target cells. Apparently, the role of exosomes in eliciting various immune responses and causing tissue injury in COVID-19 pathogenesis has been discussed. In addition, the potential of exosomes as theranostic and therapeutic agents for the treatment of COVID-19 has been elaborated.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Ben May Department for Cancer Research, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
37
|
Abbasi-Dokht T, Vafaeinezhad A, Khalesi N, Malek F, Haghmorad D, Baharlou R. T-Cell Immune Responses and Immunological Factors Associated with Coronavirus Disease 2019 Progression as Predictors for the Severity of the Disease in Hospitalized Patients. Int Arch Allergy Immunol 2023:1-10. [PMID: 36889300 PMCID: PMC10025366 DOI: 10.1159/000529513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/31/2023] [Indexed: 03/10/2023] Open
Abstract
INTRODUCTION The prevalence of coronavirus disease 2019 (COVID-19) has rapidly increased worldwide. More investigation is needed to progress toward understanding the exact role of immune responses in the pathology of the disease, leading to improved anticipation and treatment options. METHODS In the present study, we examined the relative expression of T-bet, GATA3, RORγt, and FoxP3 transcription factors as well as laboratory indicators in 79 hospitalized patients along with 20 healthy subjects as a control group. In order to make an exact comparison between various degrees of severity of disease, patients were divided into critical (n = 12) and severe (n = 67) groups. To evaluate the expression of genes of interest by performing real-time PCR, blood samples were obtained from each participant. RESULTS We found a significant increase in the expression of T-bet, GATA3, and RORγt and a reduction in the expression of FoxP3 in the critically ill patients compared to the severe and control groups. Also, we noticed that the GATA3 and RORγt expressions were elevated in the severe group in comparison with healthy subjects. Additionally, the GATA3 and RORγt expressions showed a positive correlation with elevation in CRP and hepatic enzyme concentration. Moreover, we observed that the GATA3 and RORγt expressions were the independent risk factors for the severity and outcome of COVID-19. DISCUSSION The present study showed that the overexpression of T-bet, GATA3, and RORγt, as well as a decrease in the FoxP3 expression was associated with the severity and fatal outcome of COVID-19.
Collapse
Affiliation(s)
- Tannaz Abbasi-Dokht
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Arefe Vafaeinezhad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Negin Khalesi
- Department of Internal Medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Farhad Malek
- Department of Internal Medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- *Rasoul Baharlou,
| |
Collapse
|
38
|
Berardicurti O, Navarini L, Vomero M, Currado D, Biaggi A, Marino A, Bearzi P, Corberi E, Rigon A, Arcarese L, Vadacca M, Mattei A, Agrò FE, Incalzi RA, Sambuco F, Travaglino F, Ruscitti P, Cipriani P, Giacomelli R. The similar expression of both ferritin and scavenger receptors activation genes in patients with COVID19 and AOSD support their role in the pathogenesis of these diseases and identify a common mechanism at the basis of the "hyperferritinemic syndromes". Autoimmun Rev 2023; 22:103309. [PMID: 36889656 PMCID: PMC9986116 DOI: 10.1016/j.autrev.2023.103309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
A role for COVID19 in "hyperferritinemic syndromes" has been proposed based on its clinical and serological characteristics and its similarities with AOSD. To better understand the molecular pathways responsible of these similarities, we evaluated in the PBMCs of 4 active AOSD patients, 2 COVID19 patients with ARDS, and 2 HCs the expression of genes associated with iron metabolisms, with monocyte/macrophages activation, and finally with NETs formation.
Collapse
Affiliation(s)
- Onorina Berardicurti
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy.
| | - Luca Navarini
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Marta Vomero
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Damiano Currado
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Amelia Rigon
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Luisa Arcarese
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Marta Vadacca
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Alessia Mattei
- Operative Research Unit of Anaesthesia, Intensive Care and Pain Management, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Felice Eugenio Agrò
- Operative Research Unit of Anaesthesia, Intensive Care and Pain Management, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Anesthesia, Intensive Care and Pain Management, Department of Medicine, University of Rome "Campus Bio-Medico", Rome, Italy
| | - Raffaele Antonelli Incalzi
- Unit of Geriatrics, Campus Bio-Medico University of Rome, Rome, Italy; Internal Medicine, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Federica Sambuco
- Emergency Department, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Francesco Travaglino
- Emergency Department, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| |
Collapse
|
39
|
Baseline Plasma Osteopontin Protein Elevation Predicts Adverse Outcomes in Hospitalized COVID-19 Patients. Viruses 2023; 15:v15030630. [PMID: 36992339 PMCID: PMC10054745 DOI: 10.3390/v15030630] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
More than three years have passed since the first case, and COVID-19 is still a health concern, with several open issues such as the lack of reliable predictors of a patient’s outcome. Osteopontin (OPN) is involved in inflammatory response to infection and in thrombosis driven by chronic inflammation, thus being a potential biomarker for COVID-19. The aim of the study was to evaluate OPN for predicting negative (death or need of ICU admission) or positive (discharge and/or clinical resolution within the first 14 days of hospitalization) outcome. We enrolled 133 hospitalized, moderate-to-severe COVID-19 patients in a prospective observational study between January and May 2021. Circulating OPN levels were measured by ELISA at admission and at day 7. The results showed a significant correlation between higher plasma concentrations of OPN at hospital admission and a worsening clinical condition. At multivariate analysis, after correction for demographic (age and gender) and variables of disease severity (NEWS2 and PiO2/FiO2), OPN measured at baseline predicted an adverse prognosis with an odds ratio of 1.01 (C.I. 1.0–1.01). At ROC curve analysis, baseline OPN levels higher than 437 ng/mL predicted a severe disease evolution with 53% sensitivity and 83% specificity (area under the curve 0.649, p = 0.011, likelihood ratio of 1.76, (95% confidence interval (CI): 1.35–2.28)). Our data show that OPN levels determined at the admission to hospital wards might represent a promising biomarker for early stratification of patients’ COVID-19 severity. Taken together, these results highlight the involvement of OPN in COVID-19 evolution, especially in dysregulated immune response conditions, and the possible use of OPN measurements as a prognostic tool in COVID-19.
Collapse
|
40
|
Comparison of Two Commercially Available Interferon-γ Release Assays for T-Cell-Mediated Immunity and Evaluation of Humoral Immunity against SARS-CoV-2 in Healthcare Workers. Diagnostics (Basel) 2023; 13:diagnostics13040637. [PMID: 36832126 PMCID: PMC9955378 DOI: 10.3390/diagnostics13040637] [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: 01/06/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Cellular immunity against SARS-CoV-2 is an important component of the immune response to the virus. At present, two such tests based on interferon-gamma release (interferon-γ release assays, IGRAs) are available-Quan-T-Cell SARS-CoV-2 by EUROIMMUN and T-SPOT.COVID by Oxford Immunotec. In this paper, we compared the results of these two tests in 90 subjects employed at the Public Health Institute Ostrava who had previously undergone COVID-19 infection or were vaccinated against that disease. To the best of our knowledge, this is the first head-to-head comparison of these two tests evaluating T-cell-mediated immunity against SARS-CoV-2. In addition, we also evaluated humoral immunity in the same individuals using the in-house virus neutralization test and IgG ELISA assay. The evaluation yielded similar results for both IGRAs, with Quan-T-Cell appearing to be insignificantly (p = 0.08) more sensitive (all 90 individuals were at least borderline positive) than T-SPOT.COVID (negative results found in five patients). The overall qualitative (presence/absence of immune response) agreement of both tests with virus neutralization test and anti-S IgG was also excellent (close or equal to 100% in all subgroups, with the exception of unvaccinated Omicron convalescents, a large proportion of whom, i.e., four out of six subjects, were IgG negative while at least borderline positive for T-cell-mediated immunity measured by Quan-T). This implies that the evaluation of T-cell-mediated immunity is a more sensitive indicator of immune response than the evaluation of IgG seropositivity. This is true at least for unvaccinated patients with a history of being infected only by the Omicron variant, but also likely for other groups of patients.
Collapse
|
41
|
Sameh M, Khalaf HM, Anwar AM, Osama A, Ahmed EA, Mahgoub S, Ezzeldin S, Tanios A, Alfishawy M, Said AF, Mohamed MS, Sayed AA, Magdeldin S. Integrated multiomics analysis to infer COVID-19 biological insights. Sci Rep 2023; 13:1802. [PMID: 36720931 PMCID: PMC9888750 DOI: 10.1038/s41598-023-28816-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Three years after the pandemic, we still have an imprecise comprehension of the pathogen landscape and we are left with an urgent need for early detection methods and effective therapy for severe COVID-19 patients. The implications of infection go beyond pulmonary damage since the virus hijacks the host's cellular machinery and consumes its resources. Here, we profiled the plasma proteome and metabolome of a cohort of 57 control and severe COVID-19 cases using high-resolution mass spectrometry. We analyzed their proteome and metabolome profiles with multiple depths and methodologies as conventional single omics analysis and other multi-omics integrative methods to obtain the most comprehensive method that portrays an in-depth molecular landscape of the disease. Our findings revealed that integrating the knowledge-based and statistical-based techniques (knowledge-statistical network) outperformed other methods not only on the pathway detection level but even on the number of features detected within pathways. The versatile usage of this approach could provide us with a better understanding of the molecular mechanisms behind any biological system and provide multi-dimensional therapeutic solutions by simultaneously targeting more than one pathogenic factor.
Collapse
Affiliation(s)
- Mahmoud Sameh
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Hossam M Khalaf
- Intensive Care Unit, As-Salam International Hospital, Cairo, Egypt
| | - Ali Mostafa Anwar
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Aya Osama
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Eman Ali Ahmed
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Sebaey Mahgoub
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Shahd Ezzeldin
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Anthony Tanios
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt
| | - Mostafa Alfishawy
- Infectious Diseases Consultants and Academic Researchers of Egypt (IDCARE), Cairo, Egypt
- Alazhar Center for Allergy and Immunology, Cairo, Egypt
| | - Azza Farag Said
- Department of Pulmonary Medicine, Faculty of Medicine, Minia University, Minia, Egypt
| | - Maged Salah Mohamed
- Department of Anesthesia and Intensive Care, Kasr Al Ainy, Cairo University, Cairo, Egypt
| | - Ahmed A Sayed
- Department of Basic Research, Genomics Program, Children's Cancer Hospital 57357, Cairo, Egypt
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Sameh Magdeldin
- Basic Research Department, Proteomics and Metabolomics Research Program, Children's Cancer Hospital 57357 (CCHE-57357), Cairo, Egypt.
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
42
|
Elmekaty EZI, Maklad A, Abouelhassan R, Munir W, Ibrahim MIM, Nair A, Alibrahim R, Iqbal F, Al Bishawi A, Abdelmajid A, Aboukamar M, Hadi HA, Khattab MA, Al Soub H, Al Maslamani M. Evaluation of anakinra in the management of patients with COVID-19 infection: A randomized clinical trial. Front Microbiol 2023; 14:1098703. [PMID: 36778864 PMCID: PMC9910697 DOI: 10.3389/fmicb.2023.1098703] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Background The global COVID-19 pandemic led to substantial clinical and economic outcomes with catastrophic consequences. While the majority of cases has mild to moderate disease, minority of patients progress into severe disease secondary to the stimulation of the immune response. The hyperinflammatory state contributes towards progression into multi-organ failure which necessitates suppressive therapy with variable outcomes. This study aims to explore the safety and efficacy of anakinra in COVID-19 patients with severe disease leading to cytokine release syndromes. Methods In this open-label, multi-center, randomized clinical trial, patients with confirmed COVID-19 infection with evidence of respiratory distress and signs of cytokine release syndrome were randomized in 1:1 ratio to receive either standard of care (SOC) or anakinra (100 mg subcutaneously every 12 h for 3 days then 100 mg subcutaneously once daily for 4 days) in addition to SOC. The primary outcome was treatment success at day 14 as defined by the WHO clinical progression score of ≤3. Primary analysis was based upon intention-to-treat population, with value of p of <0.05. Results Out 327 patients screened for eligibility, 80 patients were recruited for the study. The mean age was 49.9 years (SD = 11.7), with male predominance at 82.5% (n = 66). The primary outcome was not statistically different (87.5% (n = 35) in anakinra group vs. 92.5% (n = 37) in SOC group, p = 0.712; OR = 1.762 (95%CI: 0.39-7.93). The majority of reported adverse events were mild in severity and not related to the study treatment. Elevated aspartate aminotransferase was the only significant adverse event which was not associated with discontinuation of therapy. Conclusion In patients with severe COVID-19 infection, the addition of anakinra to SOC treatment was safe but was not associated with significant improvement according to the WHO clinical progression scale. Further studies are warranted to explore patients' subgroups characteristics that might benefit from administered therapy. Clinical Trial Registration Trial registration at ClinicalTrials.gov, identifier: NCT04643678.
Collapse
Affiliation(s)
- Eman Zeyad I. Elmekaty
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar,*Correspondence: Eman Zeyad I. Elmekaty,
| | - Aya Maklad
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | | | - Waqar Munir
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Arun Nair
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Rim Alibrahim
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Iqbal
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Ahmad Al Bishawi
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Mohamed Aboukamar
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Hamad Abdel Hadi
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Hussam Al Soub
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Muna Al Maslamani
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
43
|
Zhao D, Chen X, Wang L, Zhang J, Lv R, Tan L, Chen Y, Tao R, Li X, Chen Y, He W, He J. Improvement influenza vaccine immune responses with traditional Chinese medicine and its active ingredients. Front Microbiol 2023; 14:1111886. [PMID: 36960292 PMCID: PMC10027775 DOI: 10.3389/fmicb.2023.1111886] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
The current influenza vaccines are unable to provide effective protection in many cases, like influenza viruses strain antigenic drift or shift, and the influenza continues to cause significant annual morbidity and mortality. Improving the immune response to influenza vaccination is an unmet need. Traditional Chinese medicine (TCM) and its active ingredients are commonly known to have immunomodulatory properties. We therefore compared influenza vaccination alone or formulated with Astragali Radix (Huangqi in Chinese), and several representative ingredients of TCM, including lentinan (polysaccharide), panax notoginseng saponins (saponin), breviscapine (flavone), andrographolide (terpenoid), and a Chinese herbal compound (kangai) for their potential to enhance immune responses to influenza vaccine in mice. We found that all these TCM-adjuvants were able to increase hemagglutination inhibition (HAI) antibody titers, splenocyte proliferation, splenic T cell differentiation, bone marrow dendritic cell maturity, and both Th1 and Th2 cytokine secretion of influenza vaccine to varying degrees, and that had the characteristics of no excessive inflammatory responses and bidirectional regulation simultaneously. Taken together, our findings show that Astragali Radix exerts a more comprehensive effect on vaccine immunity, on both innate and adaptive immunity. The effects of lentinan and andrographolide on adaptive immunity were more significant, while the effects of breviscapine on innate immunity were stronger, and the other two TCM adjuvants were weaker. As the first report of a comprehensive evaluation of TCM adjuvants in influenza vaccines, the results suggest that TCM and their active ingredients are good candidates for enhancing the immune response of influenza vaccines, and that suitable TCMs can be selected based on the adjuvant requirements of different vaccines.
Collapse
Affiliation(s)
- Danping Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuhong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Linyuan Wang, ; Jianjun Zhang,
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Linyuan Wang, ; Jianjun Zhang,
| | - Ruilin Lv
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lingyun Tan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yawen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yan Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei He
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jing He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Kala M, Ahmad S, Dhebane M, Das K, Raturi M, Tyagi M, Kusum A. A Cross-Sectional Comparative Characterization of Hematological Changes in Patients with COVID-19 Infection, Non-COVID Influenza-like Illnesses and Healthy Controls. Viruses 2022; 15:134. [PMID: 36680172 PMCID: PMC9866193 DOI: 10.3390/v15010134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Studies have documented the role of the "neutrophil-to-lymphocyte ratio" (NLR) in influenza virus infection. In addition, morphometric parameters derived from automated analyzers on the volume, scatter and conductivity of monocytes, neutrophils and lymphocytes in many viral etiologies have helped with their early differentiation. With this background, we aimed to characterize the hematological changes of coronavirus-positive cases and also compare them with the healthy controls and patients affected by non-COVID Influenza-like illnesses so that early isolation could be considered. MATERIAL AND METHODS This was a cross-sectional analytical study carried out in the years 2020-2022. All cases with COVID-19 and non-COVID-19 Influenza-like illnesses and healthy controls above 18 years were included. Cases were diagnosed according to the WHO guidelines. All samples were processed on a Unicel DxH 800 (Beckman Coulter, California, USA) automated hematology analyzer. The demographic, clinical and regular hematological parameters along with additional parameters such as volume, conductivity and scatter (VCS) of the three groups were compared. RESULTS The 169 COVID-19 cases were in the moderate to severe category. Compared with 140 healthy controls, the majority of the routine hematological values including the NLR (neutrophil-to-lymphocyte ratio) and PLR (platelet-to-lymphocyte ratio) showed statistically significant differences. A cutoff of an absolute neutrophil count of 4350 cell/cumm was found to have a sensitivity of 76% and specificity of 70% in differentiating moderate and severe COVID-19 cases from healthy controls. COVID-19 and the non-COVID-19 Influenza-like illnesses were similar statistically in all parameters except the PLR, mean neutrophilic and monocytic volume, scatter parameters in neutrophils, axial light loss in monocytes and NLR. Interestingly, there was a trend of higher mean volumes and scatter in neutrophils and monocytes in COVID-19 cases as compared to non-COVID-19 Influenza-like illnesses. CONCLUSION We demonstrated morphological changes in neutrophils, monocytes and lymphocytes in COVID-19 infection and also non-COVID-19 Influenza-like illnesses with the help of VCS parameters. A cutoff for the absolute neutrophils count was able to differentiate COVID-19 infection requiring hospitalization from healthy controls and eosinopenia was a characteristic finding in cases with COVID-19 infection.
Collapse
Affiliation(s)
- Mansi Kala
- Department of Pathology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Sohaib Ahmad
- Department of Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Meghali Dhebane
- Department of Pathology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Kunal Das
- Department of Pediatrics, Division of Pediatric Oncology and Bone Marrow Transplantation, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Manish Raturi
- Department of Immunohematology and Blood Transfusion, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Meghna Tyagi
- Department of Pathology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Anuradha Kusum
- Department of Pathology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun 248016, Uttarakhand, India
| |
Collapse
|
45
|
Yue Y, Ma W, Accorsi EK, Ding M, Hu F, Willett WC, Chan AT, Sun Q, Rich-Edwards J, Smith-Warner SA, Bhupathiraju SN. Long-term diet and risk of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and Coronavirus Disease 2019 (COVID-19) severity. Am J Clin Nutr 2022; 116:1672-1681. [PMID: 35945354 PMCID: PMC9384672 DOI: 10.1093/ajcn/nqac219] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/04/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The role of diet on Coronavirus Disease 2019 (COVID-19) is emerging. We investigated the association between usual diet before the onset of the pandemic and risk and severity of subsequent SARS-CoV-2 infection. METHODS We included 42,935 participants aged 55-99 y in 2 ongoing cohort studies, the Nurses' Health Study II and Health Professionals Follow-up Study, who completed a series of COVID-19 surveys in 2020 and 2021. Using data from FFQs before COVID-19, we assessed diet quality using the Alternative Healthy Eating Index (AHEI)-2010, the alternative Mediterranean Diet (AMED) score, an Empirical Dietary Index for Hyperinsulinemia (EDIH), and an Empirical Dietary Inflammatory Pattern (EDIP). We calculated multivariable-adjusted ORs and 95% CIs for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and severity of COVID-19 after controlling for demographic, medical, and lifestyle factors. RESULTS Among 19,754 participants tested for SARS-CoV-2, 1941 participants reported a positive result. Of these, 1327 reported symptoms needing assistance and another 109 were hospitalized. Healthier diets, represented by higher AHEI-2010 and AMED scores and lower EDIH and EDIP scores, were associated with lower likelihood of SARS-CoV-2 infection (quartile 4 compared with quartile 1: OR: 0.80; 95% CI: 0.69, 0.92 for AHEI-2010; OR: 0.78; 95% CI: 0.67, 0.92 for AMED; OR: 1.36; 95% CI: 1.16, 1.57 for EDIH; and OR: 1.13; 95% CI: 0.99, 1.30 for EDIP; all P-trend ≤ 0.01). In the analysis of COVID-19 severity, participants with healthier diet had lower likelihood of severe infection and were less likely to be hospitalized owing to COVID-19. However, associations were no longer significant after controlling for BMI and pre-existing medical conditions. CONCLUSIONS Diet may be an important modifiable risk factor for SARS-CoV-2 infection, as well as for severity of COVID-19. This association is partially mediated by BMI and pre-existing medical conditions.
Collapse
Affiliation(s)
- Yiyang Yue
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emma K Accorsi
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Ming Ding
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Frank Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Janet Rich-Edwards
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie A Smith-Warner
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Shilpa N Bhupathiraju
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Miraglia Del Giudice M, Indolfi C, Dinardo G, Decimo F, Decimo A, Klain A. Vitamin D status can affect COVID-19 outcomes also in pediatric population. PHARMANUTRITION 2022; 22:100319. [PMID: 36268528 PMCID: PMC9562619 DOI: 10.1016/j.phanu.2022.100319] [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: 05/03/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/03/2022]
Abstract
Background vitamin D influences the immune system and the inflammatory response. It is known that vitamin D supplementation reduces the risk of acute respiratory tract infection. In the last two years, many researchers have investigated vitamin D's role in the pathophysiology of COVID-19 disease. Results the findings obtained from clinical trials and systematic reviews highlight that most patients with COVID-19 have decreased vitamin D levels and low levels of vitamin D increase the risk of severe disease. This evidence seems to be also confirmed in the pediatric population. Conclusions further studies (systematic review and meta-analysis) conducted on children are needed to confirm that vitamin D affects COVID-19 outcomes and to determine the effectiveness of supplementation and the appropriate dose, duration and mode of administration.
Collapse
Key Words
- , IFN-γ, reduce interferon-gamma
- , RAS, renin-angiotensin system
- ACE2, angiotensin-converting enzyme 2
- CI, confidence interval
- COVID-19
- Children
- DAMPs, damage-associated molecular patterns
- DCs, dendritic cells
- HR, Hazard Risk
- ICU, intensive care unit
- IL, interleukin
- IgE, immunoglobulin E
- Immunity
- MD, mean difference
- NK, natural killer
- OR, odds ratio
- PAMPs, pathogen-associated molecular patterns
- PRRs, pattern recognition receptors
- RCTs, randomized control trials
- RR, risk ratio
- SARS-CoV-2 infection
- TLRs, Toll-like receptors
- TNF-α, tumor necrosis factor-α
- Treg cells, CD, regulatory T cellcluster of differentiation
- VDRs, vitamin D receptors
- Vitamin D
Collapse
Affiliation(s)
- Michele Miraglia Del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Cristiana Indolfi
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giulio Dinardo
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabio Decimo
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alberto Decimo
- Department of Mental, Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Angela Klain
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
47
|
Nazerian Y, Ghasemi M, Yassaghi Y, Nazerian A, Mahmoud Hashemi S. Role of SARS-CoV-2-induced Cytokine Storm in Multi-Organ Failure: Molecular Pathways and Potential Therapeutic Options. Int Immunopharmacol 2022; 113:109428. [PMCID: PMC9637536 DOI: 10.1016/j.intimp.2022.109428] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
Coronavirus disease 2019 (COVID-19) outbreak has become a global public health emergency and has led to devastating results. Mounting evidence proposes that the disease causes severe pulmonary involvement and influences different organs, leading to a critical situation named multi-organ failure. It is yet to be fully clarified how the disease becomes so deadly in some patients. However, it is proven that a condition called “cytokine storm” is involved in the deterioration of COVID-19. Although beneficial, sustained production of cytokines and overabundance of inflammatory mediators causing cytokine storm can lead to collateral vital organ damages. Furthermore, cytokine storm can cause post-COVID-19 syndrome (PCS), an important cause of morbidity after the acute phase of COVID-19. Herein, we aim to explain the possible pathophysiology mechanisms involved in COVID-19-related cytokine storm and its association with multi-organ failure and PCS. We also discuss the latest advances in finding the potential therapeutic targets to control cytokine storm wishing to answer unmet clinical demands for treatment of COVID-19.
Collapse
Affiliation(s)
- Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Ghasemi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Younes Yassaghi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Seyed Mahmoud Hashemi
- Medical nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding author at: Medical nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran / Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Hastak PS, Andersen CR, Kelleher AD, Sasson SC. Frontline workers: Mediators of mucosal immunity in community acquired pneumonia and COVID-19. Front Immunol 2022; 13:983550. [PMID: 36211412 PMCID: PMC9539803 DOI: 10.3389/fimmu.2022.983550] [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: 07/01/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
The current COVID-19 pandemic has highlighted a need to further understand lung mucosal immunity to reduce the burden of community acquired pneumonia, including that caused by the SARS-CoV-2 virus. Local mucosal immunity provides the first line of defence against respiratory pathogens, however very little is known about the mechanisms involved, with a majority of literature on respiratory infections based on the examination of peripheral blood. The mortality for severe community acquired pneumonia has been rising annually, even prior to the current pandemic, highlighting a significant need to increase knowledge, understanding and research in this field. In this review we profile key mediators of lung mucosal immunity, the dysfunction that occurs in the diseased lung microenvironment including the imbalance of inflammatory mediators and dysbiosis of the local microbiome. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality.
Collapse
Affiliation(s)
- Priyanka S. Hastak
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Christopher R. Andersen
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
- Intensive Care Unit, Royal North Shore Hospital, Sydney, NSW, Australia
- Critical Care and Trauma Division, The George Institute for Global Health, Sydney, NSW, Australia
| | - Anthony D. Kelleher
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Sarah C. Sasson
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
49
|
Ji XS, Chen B, Ze B, Zhou WH. Human genetic basis of severe or critical illness in COVID-19. Front Cell Infect Microbiol 2022; 12:963239. [PMID: 36204639 PMCID: PMC9530247 DOI: 10.3389/fcimb.2022.963239] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to considerable morbidity and mortality worldwide. The clinical manifestation of COVID-19 ranges from asymptomatic or mild infection to severe or critical illness, such as respiratory failure, multi-organ dysfunction or even death. Large-scale genetic association studies have indicated that genetic variations affecting SARS-CoV-2 receptors (angiotensin-converting enzymes, transmembrane serine protease-2) and immune components (Interferons, Interleukins, Toll-like receptors and Human leukocyte antigen) are critical host determinants related to the severity of COVID-19. Genetic background, such as 3p21.31 and 9q34.2 loci were also identified to influence outcomes of COVID-19. In this review, we aimed to summarize the current literature focusing on human genetic factors that may contribute to the observed diversified severity of COVID-19. Enhanced understanding of host genetic factors and viral interactions of SARS-CoV-2 could provide scientific bases for personalized preventive measures and precision medicine strategies.
Collapse
Affiliation(s)
- Xiao-Shan Ji
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| | - Bin Chen
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| | - Bi Ze
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| |
Collapse
|
50
|
Singhal P, Rawat A, Sharma S, Sharma AK, Jat KS, Agarwal S, Samdani S, Agarwal S, Sharma MP, Bhandari S. Invasive Fungal Infections in Dengue Recovered Patients: A New Phenomenon in the Covid-19 Era. Indian J Otolaryngol Head Neck Surg 2022; 74:3321-3326. [PMID: 36090297 PMCID: PMC9441130 DOI: 10.1007/s12070-022-03152-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/25/2022] [Indexed: 11/26/2022] Open
Abstract
Invasive fungal rhinosinusitis was seen to rise to epidemic levels after the 2nd wave of ongoing Covid pandemic, especially in tropical countries, maximally in India. A similar trend is being observed for cases who have recently recovered from dengue virus infection. Post dengue invasive fungal infection is a new presentation and any associations between it and Covid pandemic need to be studied in detail to help prepare for any complications. 3 patients presented to the out-patient department of E.N.T at a tertiary level teaching hospital in East India with complains similar to rhinosinusitis. These patients were then evaluated and diagnosed to be infected from Mucormycosis and Aspergillosis fungal sinusitis after which they were managed with surgical debridement and systemic antifungal therapy. All had a recent history of recovery from Dengue virus infection and a possible association could be suspected. 3 patients presented with complains of pain over upper jaw with orbital swelling and loss of vision developing over a period of 24 days. Two of them had ulceration of hard palate. They were then subjected to Contrast MRI along with CT scan of the Paranasal sinuses which depicted pansinus involvement with intracranial extension in two patients. These were then planned for diagnostic nasal endoscopies along with biopsies which turned out to be Invasive fungal sinusitis in the form of Aspergillosis and Mucormycosis. All the 3 patients had recent history of recovery from Dengue virus infection and did not have any other co-morbidities. Covid Associated Mucormycosis (CAM) is a well-known entity now but no reports of Dengue associated Invasive fungal sinusitis are yet reported. Whether this new phenomenon has anything to do with the interactions between dengue virus and coronavirus is not known at present and needs to be studied in detail so appropriate management protocols can be formulated.
Collapse
Affiliation(s)
- Pawan Singhal
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Anshu Rawat
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Shivam Sharma
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Anjani Kumar Sharma
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Kailash Singh Jat
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Shubham Agarwal
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Sunil Samdani
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Sunita Agarwal
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Man Prakash Sharma
- Department of Otorhinolaryngology and Head Neck Surgery, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| | - Sudheer Bhandari
- Department of Internal Medicine, S.M.S Medical College and Hospital, Jaipur, Rajasthan 302004 India
| |
Collapse
|