1
|
Buvvaji SM, Joshi V, Angel A, Angel B, Khaneja P, Joshi R. Intracellular alterations, vacuolization and bypass mechanism by SARS-CoV-2 infection could be the possible basis of respiratory distress and hypoxia. Tissue Cell 2025; 95:102896. [PMID: 40188686 DOI: 10.1016/j.tice.2025.102896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 causes mild to severe Acute Respiratory Distress Syndrome, Pneumonia and lung tissue damage. This leads to sub performance in the pulmonary gaseous exchange by the alveolar cells causing hypoxia associated with clinical severities/mortality. The exact cellular basis of the pulmonary malfunction resulting into death of approximately 7.1 million people needs to be fully studied. Understanding the intracellular alterations in pulmonary cells caused by viral infection could prove to be a significant step in our attempts to revert the respiratory efficiency of the patients through appropriate therapeutic interventions. We have undertaken In-Vitro studies to understand the pathogenesis of SARS-CoV-2 in alveoli. We cultured the Alveolar Epithelium (A549 and L-132), Fibroblasts (WI-38), Human Pulmonary Artery Endothelial Cells (HPAEC-c), and African Green Monkey Kidney Epithelial Cells (Vero-E6) and infected them with SARS-CoV-2. Vacuoles in infected Alveolar Type-2 cells, cytoskeletal deformation, fragmentation of mitochondria in alveolar and arterial endothelial cells, loss of glycoclayx in endothelial cells and a unique bypass exit mechanism of virus were observed as major intracellular changes due to infection. The bypass exit of the daughter virions from lung cells along with loss of glycoclayx due to virus overburdening is reported as mechanism of propagation of infection towards multiple organs. We report that formation of numerous vacuoles in infected Alveolar Type-2 cells and the SARS-CoV-2 virions occupying these vacuoles could hamper the trans cytoplasmic trafficking of surfactant mixed inspired air and its subsequent transfer into venous blood through cell membranes of Alveolar Type -2 Cells and Capillary Wall Cells of pulmonary vein. The possible use of repurposed Nitroglycerine based drug to retrieve required intracellular cytoplasmic viscosity of the Alveolar type 2 cells has also been suggested.
Collapse
Affiliation(s)
- Shareef Mohammed Buvvaji
- Centre of Excellence in Virology & Immunology, Sharda University, Greater Noida, U.P. 201310, India
| | - Vinod Joshi
- Centre of Excellence in Virology & Immunology, Sharda University, Greater Noida, U.P. 201310, India.
| | - Annette Angel
- Centre of Excellence in Virology & Immunology, Sharda University, Greater Noida, U.P. 201310, India
| | - Bennet Angel
- Centre of Excellence in Virology & Immunology, Sharda University, Greater Noida, U.P. 201310, India
| | - Poorna Khaneja
- Centre of Excellence in Virology & Immunology, Sharda University, Greater Noida, U.P. 201310, India
| | - Ramesh Joshi
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, U.P. 201310, India
| |
Collapse
|
2
|
Wang Y, Cheng Y, Wang S, Liu D, Gao Y, Li J, Jiang Y, Cui W, Qiao X, Li Y, Wang L. Unraveling the cross-talk between a highly virulent PEDV strain and the host via single-cell transcriptomic analysis. J Virol 2025:e0055525. [PMID: 40396761 DOI: 10.1128/jvi.00555-25] [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: 03/26/2025] [Accepted: 04/28/2025] [Indexed: 05/22/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe intestinal damage and high mortality in neonatal piglets. The continuous emergence of new strains has brought new challenges to prevention and control. In this study, we isolated and characterized a prevalent PEDV virulent strain and analyzed 19,612 jejunal cells from PEDV-infected and control piglets using single-cell sequencing, revealing significant changes in cellular composition, gene expression, and intercellular communication. In response to PEDV infection, epithelial repair was enhanced through increased proliferation and differentiation of stem cells, transit-amplifying (TA) cells, and intestinal progenitor cells into enterocytes. Additionally, PEDV disrupted intercellular communication, compromising epithelial functionality while triggering immune responses, with IFN-γ and IL-10 signaling activation acting as critical regulators of immune balance and tissue homeostasis. Beyond enterocytes, viral genes were detected in various other cell types. Further experiments confirmed that PEDV could initiate replication in B and T lymphocytes but was unable to produce infectious progeny, with T cells additionally undergoing virus-induced apoptosis. These findings provide new insights into PEDV tropism, immune evasion, and epithelial repair, revealing complex host-pathogen interactions that shape disease progression and tissue regeneration, thereby contributing to a better understanding of enteric coronavirus pathogenesis.IMPORTANCEThe persistent circulation of porcine epidemic diarrhea virus (PEDV) poses a major threat to the swine industry, with emerging strains complicating prevention and control efforts. Currently, no effective measures completely prevent virus transmission, highlighting the need to understand PEDV-host interactions. In this study, we isolated a prevalent virulent strain and used single-cell sequencing to identify new PEDV-infected cell types and explore the complex interplay between the host and PEDV. These findings provide essential insights into viral pathogenesis and facilitate the design of targeted antiviral interventions.
Collapse
Affiliation(s)
- Yanan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yu Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing, China
| | - Yueyi Gao
- China Institute of Veterinary Drug Control, Beijing, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
3
|
Huang S, Zhou Y, Ji H, Zhang T, Liu S, Ma L, Deng D, Ding Y, Han L, Shu S, Wang Y, Chen X. Decoding mechanisms and protein markers in lung-brain axis. Respir Res 2025; 26:190. [PMID: 40390067 DOI: 10.1186/s12931-025-03272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 05/08/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The lung-brain axis represents a complex bidirectional communication network that is pivotal in the crosstalk between respiratory and neurological functions. This review summarizes the current understanding of the mechanisms and protein markers that mediate the effects of lung diseases on brain health. MAIN FINDINGS In this review, we explore the mechanisms linking lung injury to neurocognitive impairments, focusing on neural pathways, immune regulation and inflammatory responses, microorganism pathways, and hypoxemia. Specifically, we highlight the role of the vagus nerve in modulating the central nervous system response to pulmonary stimuli; Additionally, the regulatory function of the immune system is underscored, with evidence suggesting that lung-derived immune mediators can traverse the blood-brain barrier, induce neuroinflammation and cognitive decline; Furthermore, we discuss the potential of lung microbiota to influence brain diseases through microbial translocation and immune activation; Finally, the impact of hypoxemia is examined, with findings indicating that it can exacerbate cerebral injury via oxidative stress and impaired perfusion. Moreover, we analyze how pulmonary conditions, such as pneumonia, ALI/ARDS, and asthma, contribute to neurological dysfunction. Prolonged mechanical ventilation can also contribute to cognitive impairment. Conversely, brain diseases (e.g., stroke, traumatic brain injury) can lead to acute respiratory complications. In addition, protein markers such as TLR4, ACE2, A-SAA, HMGB1, and TREM2 are crucial to the lung-brain axis and correlate with disease severity. We also discuss emerging therapeutic strategies targeting this axis, including immunomodulation and microbiome engineering. Overall, understanding the lung-brain interplay is crucial for developing integrated treatment strategies and improving patient outcomes. Further research is needed to elucidate the molecular mechanisms and foster interdisciplinary collaboration.
Collapse
Affiliation(s)
- Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuxi Zhou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Haipeng Ji
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shiya Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Lulin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Shaofang Shu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China
| | - Yu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Wuhan, 430022, China.
| |
Collapse
|
4
|
El-Naas A, Hamad O, Nair S, Alfakhri B, Mahmoud S, Haji A, Ahmed L, Lebbe A, Aboulwafa A, Shaikh F, Bouhali I, Zakaria D. New Onset of Type 1 and Type 2 Diabetes Post-COVID-19 Infection: A Systematic Review. Emerg Microbes Infect 2025:2492211. [PMID: 40326310 DOI: 10.1080/22221751.2025.2492211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
AbstractCOVID-19 may primarily cause respiratory symptoms but can lead to long-term effects known as long COVID. COVID-19-induced diabetes mellitus was reported in many patients which shares characteristics of types 1 and 2 (T1DM and T2DM). This study aims to identify and analyze the reported cases of new onset diabetes post-COVID-19 infection. Several databases were used to conduct a comprehensive literature search to target studies reporting cases of T1DM or T2DM post-COVID-19 infection. Screening, data extraction, and cross checking were performed by two independent reviewers. Only 43 studies met our inclusion criteria. Our results revealed that the overall prevalence of new onset diabetes post-COVID-19 was 1.37% with higher prevalence for T2DM (0.84%) as compared to T1DM (0.017%) while the type of diabetes was not reported in 0.51% of the cases. Several risk factors for developing diabetes post-COVID-19 infection were identified including the type of SARS-CoV-2 variant, age, comorbidities and the vaccination status. The direct viral attack of the pancreatic beta cells as well as inflammation and the anti-inflammatory corticosteroids were proposed as possible mechanisms of the COVID-19 induced diabetes. A multidisciplinary approach involving endocrinologists, primary care physicians, and infectious disease specialists should be implemented in the management of post-COVID patients to address both the acute and long-term complications, including metabolic changes and risk of diabetes.
Collapse
Affiliation(s)
- Ahmed El-Naas
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Omar Hamad
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Siddhant Nair
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Bushra Alfakhri
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shadi Mahmoud
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Aliyaa Haji
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Lina Ahmed
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ahamed Lebbe
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ali Aboulwafa
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Farha Shaikh
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Imane Bouhali
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Dalia Zakaria
- Department of Premedical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
5
|
Zhu Y, Gao Z, Feng X, Hu Y, Liu N, Liu C, Yang Q, Zou Q, Li M, Song G, He Y. Comprehensive preclinical characterization of IPB29, a pan-coronavirus fusion inhibitor under clinical trials. Antiviral Res 2025; 237:106154. [PMID: 40158858 DOI: 10.1016/j.antiviral.2025.106154] [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: 01/23/2025] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
IPB29 is a lipopeptide-based coronavirus fusion inhibitor with the potent, broad-spectrum antiviral activity, and it has already been advanced to phase III clinical trials for the treatment of SARS-CoV-2 infection. We recently reported its design strategy and initial preclinical characterization; herein, we focused on characterizing its efficacies against newly-emerged Omicron variants, as well as its chronic general toxicity, toxicokinetics, immunogenicity, and reproductive toxicity in animal models. As anticipated, IPB29 demonstrated improved activity in inhibiting JN.1 and KP.2 variants, effectively blocking cell fusion and pseudovirus infections. Nebulized inhalation of IPB29 exhibited high therapeutic efficacy against live BA.5 and EG.5.1 infections in Syrian hamsters. The 26-week toxicity studies revealed that nebulized IPB29 has a favorable safety profile, with well-characterized toxicokinetics in SD rats and Beagle dogs. Notably, short-term nebulization of IPB29 did not elicit anti-drug antibody (ADA) responses in either species. However, IPB29-specific antibodies were detected after long-term administration. Finally, a three-stage reproductive toxicity study in SD rats indicated that IPB29 had no significant toxic effects on fertility, embryo-fetal development, or the development of offspring. In summary, our findings demonstrate that IPB29 is a safe and effective SARS-CoV-2 inhibitor with promising potential for clinical applications.
Collapse
Affiliation(s)
- Yuanmei Zhu
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhongcai Gao
- Research Institute of Youcare Pharmaceutical Group Co., Ltd., Beijing, 100176, China
| | - Xiaoli Feng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Yue Hu
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Nian Liu
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chao Liu
- Research Institute of Youcare Pharmaceutical Group Co., Ltd., Beijing, 100176, China
| | - Qiaojiang Yang
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Qingcui Zou
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Minghua Li
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| | - Gengshen Song
- Research Institute of Youcare Pharmaceutical Group Co., Ltd., Beijing, 100176, China.
| | - Yuxian He
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
6
|
Huang X, Gao H, Zhang J, Zhan P, Liu X. A patent review of anti-coronavirus agents targeting the spike-ACE2 interaction (2019-present). Expert Opin Ther Pat 2025:1-12. [PMID: 40259874 DOI: 10.1080/13543776.2025.2494860] [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: 12/05/2024] [Revised: 02/24/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
INTRODUCTION The Angiotensin-converting enzyme 2 (ACE2) receptor, crucial for coronavirus recognition of host cells, is a key target for therapeutic intervention against SARS-CoV-2 and related coronaviruses. Therefore, thoroughly investigating the interaction mechanism between ACE2 and the Spike protein (S protein), as well as developing targeted inhibitors based on this mechanism, is vital for effectively controlling the spread of SARS-CoV-2 and preventing potential future pandemics caused by other coronaviruses. AREAS COVERED This article comprehensively reviews the mechanisms underlying ACE2-S protein interaction that facilitate SARS-CoV-2 entry into host cells. It also analyzes the patent landscape regarding inhibitors targeting the ACE2-S interface since 2019. EXPERT OPINION In the 5 years since the outbreak of SARS-CoV-2, numerous methods and design strategies have been employed to develop innovative therapeutics against coronaviruses. Among these approaches, inhibitors targeting both the ACE2 receptor and the S protein have gained significant interest due to their potential in blocking various coronaviruses. Despite facing challenges similar to other protein-protein interaction inhibitors, progress has been made in developing these inhibitors through virtual screening, covalent protein binding, and peptide modification strategies. However, obstacles persist in clinical translation, necessitating a multidisciplinary strategy that integrates state-of-the-art methodologies to optimize S-ACE2 interface-targeted drug discovery.
Collapse
Affiliation(s)
- Xing Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Heng Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
7
|
Uwamahoro H, Collier WE, Nashar TO, Jaynes JM, Mortley DG, Davis CG, Kanyairita GG, Abdelazim EF, Igiramaboko JFR, Habineza C, Tumushimiyimana D, Rutayisire UN, Davis YA, Renard KL. Natural and Designed Cyclic Peptides as Potential Antiviral Drugs to Combat Future Coronavirus Outbreaks. Molecules 2025; 30:1651. [PMID: 40333520 PMCID: PMC12029270 DOI: 10.3390/molecules30081651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 05/09/2025] Open
Abstract
The COVID-19 pandemic has underscored the need for effective and affordable antiviral drugs. Anthropogenic activities have increased interactions among humans, animals, and wildlife, contributing to the emergence of new and re-emerging viral diseases. RNA viruses pose significant challenges due to their rapid mutation rates, high transmissibility, and ability to adapt to host immune responses and antiviral treatments. The World Health Organization has identified several diseases (COVID-19, Ebola, Marburg, Zika, and others), all caused by RNA viruses, designated as being of priority concern as potential causes of future pandemics. Despite advances in antiviral treatments, many viruses lack specific therapeutic options, and more importantly, there is a paucity of broad-spectrum antiviral drugs. Additionally, the high costs of current treatments such as Remdesivir and Paxlovid highlight the need for more affordable antiviral drugs. Cyclic peptides from natural sources or designed through molecular modeling have shown promise as antiviral drugs with stability, low toxicity, high target specificity, and low antiviral resistance properties. This review emphasizes the urgent need to develop specific and broad-spectrum antiviral drugs and highlights cyclic peptides as a sustainable solution to combat future pandemics. Further research into these compounds could provide a new weapon to combat RNA viruses and address the gaps in current antiviral drug development.
Collapse
Affiliation(s)
- Hilarie Uwamahoro
- Department of Chemistry, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (H.U.); (J.M.J.); (G.G.K.); (E.F.A.)
| | - Willard E. Collier
- Department of Chemistry, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (H.U.); (J.M.J.); (G.G.K.); (E.F.A.)
| | - Toufic O. Nashar
- Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Jesse M. Jaynes
- Department of Chemistry, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (H.U.); (J.M.J.); (G.G.K.); (E.F.A.)
- Department of Agricultural and Environmental Sciences, College of Agriculture, Environment & Nutrition Sciences, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Desmond G. Mortley
- Department of Agricultural and Environmental Sciences, College of Agriculture, Environment & Nutrition Sciences, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Cheryl G. Davis
- Department of Biology, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (C.G.D.); (Y.A.D.)
| | - Getrude G. Kanyairita
- Department of Chemistry, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (H.U.); (J.M.J.); (G.G.K.); (E.F.A.)
| | - Eslam F. Abdelazim
- Department of Chemistry, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (H.U.); (J.M.J.); (G.G.K.); (E.F.A.)
| | | | - Concorde Habineza
- Computational Data Science & Engineering, College of Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA;
| | - Devotha Tumushimiyimana
- Department of Human Ecology, College of Agriculture, Science and Technology, Delaware State University, Dover, DE 19901, USA;
| | - Umuraza Noella Rutayisire
- Department of Natural Resources and Environmental Sciences, College of Agricultural, Life and Natural Sciences, Normal, AL 35811, USA;
| | - Yasmin A. Davis
- Department of Biology, College of Arts & Sciences, Tuskegee University, Tuskegee, AL 36088, USA; (C.G.D.); (Y.A.D.)
| | - Kamora L. Renard
- Department of Health Science, School of Nursing & Allied Health, Tuskegee University, Tuskegee, AL 36088, USA;
| |
Collapse
|
8
|
Emam MH, Elezaby RS, Swidan SA, Loutfy SA, Hathout RM. Enhancing Polyacrylonitrile Nanofibers Antiviral Activity Using Greenly Synthesized Silver Nanoparticles. Arch Pharm (Weinheim) 2025; 358:e202400943. [PMID: 40165662 DOI: 10.1002/ardp.202400943] [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: 12/17/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Developing efficient antiviral protectives is a new approach against respiratory emerging viruses. This study aims to synthesize silver nanoparticles (Ag NPs) via a green technique using crocin to provide a virucidal effect and to enhance the protection of polyacrylonitrile (PAN) nanofibrous face masks or respirators against viruses. The influence of formulation and process variables on the particle size (PS) of Ag NPs was studied using d-optimal response surface design. The selected NPs were loaded into PAN nanofibers (NFs). MTT colorimetric assay was performed to determine the safety of the prepared NPs and NFs on Vero cells. Further, an immunofluorescent assay was performed to determine the composite's ability to inhibit the ACE2-SARS-CoV-2 spike protein interaction and prevent viral infection. The selected NPs possessed a small PS of 23.21 ± 0.86 nm, a PDI of 0.23 ± 0.019, and a ZP of -21.8 ± 1.82 mV. The optimum NF composite was fabricated with a PAN concentration of 8% w/v loaded with 0.25% w/w Ag NPs, with a feeding rate of 0.7 mL/h and an applied voltage of 23.5 kV. The resultant NFs displayed an acceptable morphology and a mean diameter of 378.88 ± 91.12 nm. In vitro cytotoxicity studies on Vero cells revealed the biocompatibility of crocin and Ag NPs. Moreover, Ag-PAN NFs were proven biologically safe. The immunofluorescent assay showed that Ag-PAN NFs demonstrated the least IC50 value of 10.99 µg/mL, indicating their potent effect on inhibiting SARS-CoV-2 infection. Ag-PAN NFs are a promising safe antiviral composite that has the potential to be used in face masks.
Collapse
Affiliation(s)
- Merna H Emam
- Nanotechnology Research Center (NTRC), The British University in Egypt, El-Shorouk City, Cairo, Egypt
| | - Reham S Elezaby
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Shady A Swidan
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Rania M Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| |
Collapse
|
9
|
Pujadas V, Chin C, Sankpal NV, Buhrmaster J, Arjuna A, Walia R, Smith MA, Eickelberg O, Bremner RM, Mohanakumar T, Sureshbabu A. Alveolar epithelial type 2 cell specific loss of IGFBP2 activates inflammation in COVID-19. Respir Res 2025; 26:111. [PMID: 40121473 PMCID: PMC11929192 DOI: 10.1186/s12931-025-03187-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) global pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, our understanding of SARS-CoV-2-induced inflammation in alveolar epithelial cells remains very limited. The contributions of intracellular insulin-like growth factor binding protein-2 (IGFBP2) to SARS-CoV-2 pathogenesis are also unclear. In this study, we have uncovered a critical role for IGFBP2, specifically in alveolar epithelial type 2 cells (AEC2), in the immunopathogenesis of COVID-19. Using bulk RNA sequencing, we show that IGFBP2 mRNA expression is significantly downregulated in primary AEC2 cells isolated from fibrotic lung regions from patients with COVID-19-acute respiratory distress syndrome (ARDS) compared to those with idiopathic pulmonary fibrosis (IPF) alone or IPF with a history of COVID-19. Using multicolor immunohistochemistry, we demonstrated that IGFBP2 and its selective ligands IGF1 and IGF2 were significantly reduced in AEC2 cells from patients with COVID-ARDS, IPF alone, or IPF with COVID history than in those from age-matched donor controls. Further, we demonstrated that lentiviral expression of Igfbp2 significantly reduced mRNA expression of proinflammatory cytokines-Tnf-α, Il1β, Il6, Stat3, Stat6 and chemokine receptors-Ccr2 and Ccr5-in mouse lung epithelial cells challenged with SARS-CoV-2 spike protein injury (S2; 500 ng/mL). Finally, we demonstrated higher levels of cytokines-TNF-α; IL-6 and chemokine receptor-CCR5 in AEC2 cells from COVID-ARDS patients compared to the IPF alone and the IPF with COVID history patients. Altogether, these data suggest that anti-inflammatory properties of IGFBP2 in AEC2 cells and its localized delivery may serve as potential therapeutic strategy for patients with COVID-19.
Collapse
Affiliation(s)
- Valentina Pujadas
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Chiahsuan Chin
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Narendra V Sankpal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - James Buhrmaster
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Ashwini Arjuna
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Thalachallour Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA.
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA.
| |
Collapse
|
10
|
Lin MW, Lin CH, Chang JR, Chiang HH, Wu TH, Lin CS. The influence of PM2.5 exposure on SARS-CoV-2 infection via modulating the expression of angiotensin converting enzyme II. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136887. [PMID: 39700942 DOI: 10.1016/j.jhazmat.2024.136887] [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: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Particulate matter 2.5 (PM2.5) pollution and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic are the greatest environmental health issues worldwide. Several statistics revealed the significant positive correlation between the morbidity of coronavirus disease-19 (COVID-19) and the levels of air pollution. Nevertheless, there is no direct experimental evidence to indicate the effect of PM2.5 exposure on SARS-CoV-2 infection. The objective of this study was to evaluate whether the infection of SARS-CoV-2 affected by PM2.5 through angiotensin-converting enzyme II (ACE2) expression enhances and investigate the function of ACE2 in lung injury induced by PM2.5. An animal model of PM2.5-induced lung injury was established using wild-type (WT, C57BL/6), human ACE2 transgenic (K18-hACE2 TG), and murine ACE2 gene knockout (mACE2 KO) mice. The results indicate that PM2.5 exposure facilitates SARS-CoV-2 infection through inducing ACE2 expression in vitro (10 μg/mL) and in vivo (6.25 mg/kg/day in 50 μL saline). The levels of ACE, inflammatory cytokines, and mitogen-activated protein kinase (MAPK) proteins in WT, K18-hACE TG and mACE2 KO mice were significantly increased after PM2.5 instillation. The severest PM2.5-induced lung damage was observed in mACE2 KO mice. In summary, ACE2 plays a double-edged sword role in lung injury, PM2.5 exposure contributed to SARS-CoV-2 infection through inducing ACE2 expression, but ACE2 also protected pulmonary inflammation from PM2.5 challenge.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Cheng-Han Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Jia-Rong Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Hua-Hsin Chiang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Ting-Hsuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
11
|
Conca DV, Bano F, Graul M, von Wirén J, Scherrer L, Pace H, Sharma H, Svirelis J, Thorsteinsson K, Dahlin A, Bally M. Variant-Specific Interactions at the Plasma Membrane: Heparan Sulfate's Impact on SARS-CoV-2 Binding Kinetics. Anal Chem 2025; 97:4318-4328. [PMID: 39976108 PMCID: PMC11883730 DOI: 10.1021/acs.analchem.4c04283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
The spread of SARS-CoV-2 led to the emergence of several variants of concern (VOCs). The spike glycoprotein, responsible for engaging the viral receptor, exhibits the highest density of mutations, suggesting an ongoing evolution to optimize viral entry. This study characterizes the bond formed by virion mimics carrying the SARS-CoV-2 spike protein and the plasma membrane of host cells in the early stages of virus entry. Contrary to the traditional analysis of isolated ligand-receptor pairs, we utilized well-defined biomimetic models and biochemical and biophysical techniques to characterize the multivalent interaction of VOCs with the complex cell membrane. We observed an overall increase in the binding affinity for newer VOCs. By progressively reducing the system complexity, we identify heparan sulfate (HS) as a main driver of this variation, with a 10-fold increase in affinity for Omicron BA.1 over that of the original strain. These results demonstrate the essential role of coreceptors, particularly HS, in the modulation of SARS-CoV-2 infection and highlight the importance of multiscale biophysical and biochemical assays that account for membrane complexity to fully characterize and understand the role of molecular components and their synergy in viral attachment and entry.
Collapse
Affiliation(s)
- Dario Valter Conca
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Fouzia Bano
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Małgorzata Graul
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Julius von Wirén
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Lauriane Scherrer
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Hudson Pace
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Himanshu Sharma
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
- Laboratory
for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå 901 87, Sweden
| | - Justas Svirelis
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, Gothenburg 412 96, Sweden
| | - Konrad Thorsteinsson
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Andreas Dahlin
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, Gothenburg 412 96, Sweden
| | - Marta Bally
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| |
Collapse
|
12
|
Wang Y, Yang Z, Zheng X, Liang X, Wu L, Wu C, Dai J, Cao Y, Li M, Zhou F. Cerebral blood flow alterations and host genetic association in individuals with long COVID: A transcriptomic-neuroimaging study. J Cereb Blood Flow Metab 2025; 45:431-442. [PMID: 39177056 PMCID: PMC11572096 DOI: 10.1177/0271678x241277621] [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: 03/15/2024] [Revised: 07/03/2024] [Accepted: 08/03/2024] [Indexed: 08/24/2024]
Abstract
Neuroimaging studies have indicated that altered cerebral blood flow (CBF) was associated with the long-term symptoms of postacute sequelae of SARS-CoV-2 infection (PASC), also known as "long COVID". COVID-19 and long COVID were found to be strongly associated with host gene expression. Nevertheless, the relationships between altered CBF, clinical symptoms, and gene expression in the central nervous system (CNS) remain unclear in individuals with long COVID. This study aimed to explore the genetic mechanisms of CBF abnormalities in individuals with long COVID by transcriptomic-neuroimaging spatial association. Lower CBF in the left frontal-temporal gyrus was associated with higher fatigue and worse cognition in individuals with long COVID. This CBF pattern was spatially associated with the expression of 2,178 genes, which were enriched in the molecular functions and biological pathways of COVID-19. Our study suggested that lower CBF is associated with persistent clinical symptoms in long COVID individuals, possibly as a consequence of the complex interactions among multiple COVID-19-related genes, which contributes to our understanding of the impact of adverse CNS outcomes and the trajectory of development to long COVID.
Collapse
Affiliation(s)
- Yao Wang
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| | - Ziwei Yang
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| | - Xiumei Zheng
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| | - Xiao Liang
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| | - Lin Wu
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| | - Chengsi Wu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | | | - Yuan Cao
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center for Medical Imaging in Jiangxi Province, Nanchang, China
| |
Collapse
|
13
|
Tsai YX, Chien YC, Hsu MF, Khoo KH, Hsu STD. Molecular basis of host recognition of human coronavirus 229E. Nat Commun 2025; 16:2045. [PMID: 40016196 PMCID: PMC11868633 DOI: 10.1038/s41467-025-57359-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
Human coronavirus 229E (HCoV-229E) is the earliest CoV found to infect humans. It binds to the human aminopeptidase N (hAPN) through the receptor binding domain (RBD) of its spike (S) protein to achieve host recognition. We present the cryo-electron microscopy structure of two HCoV-229E S protein in complex with a dimeric hAPN to provide structural insights on how the HCoV-229E S protein opens up its RBD to engage with its host receptor, information that is currently missing among alphacoronaviruses to which HCoV-229E belong. We quantitatively profile the glycosylation of HCoV-229E S protein and hAPN to deduce the glyco-shielding effects pertinent to antigenicity and host recognition. Finally, we present an atomic model of fully glycosylated HCoV-229E S in complex with hAPN anchored on their respective membrane bilayers to recapitulate the structural basis of the first step of host infection by HCoV-229E.
Collapse
Affiliation(s)
- Yu-Xi Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Yu-Chun Chien
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM²), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| |
Collapse
|
14
|
Serra-Llovich A, Cullell N, Maroñas O, José Herrero M, Cruz R, Almoguera B, Ayuso C, López-Rodríguez R, Domínguez-Garrido E, Ortiz-Lopez R, Barreda-Sánchez M, Corton M, Dalmau D, Calbo E, Boix-Palop L, Dietl B, Sangil A, Gil-Rodriguez A, Guillén-Navarro E, Mancebo E, Lira-Albarrán S, Minguez P, Paz-Artal E, Olivera GG, Recarey-Rama S, Sendra L, Zucchet EG, López de Heredia M, Flores C, Riancho JA, Rojas-Martinez A, Lapunzina P, Carracedo Á, Arranz MJ. Pharmacogenomic Study of SARS-CoV-2 Treatments: Identifying Polymorphisms Associated with Treatment Response in COVID-19 Patients. Biomedicines 2025; 13:553. [PMID: 40149530 PMCID: PMC11940783 DOI: 10.3390/biomedicines13030553] [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: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The COVID-19 pandemic resulted in 675 million cases and 6.9 million deaths by 2022. Despite substantial declines in case fatalities following widespread vaccination campaigns, the threat of future coronavirus outbreaks remains a concern. Current treatments for COVID-19 have been repurposed from existing therapies for other infectious and non-infectious diseases. Emerging evidence suggests a role for genetic factors in both susceptibility to SARS-CoV-2 infection and response to treatment. However, comprehensive studies correlating clinical outcomes with genetic variants are lacking. The main aim of our study is the identification of host genetic biomarkers that predict the clinical outcome of COVID-19 pharmacological treatments. Methods: In this study, we present findings from GWAS and candidate gene and pathway enrichment analyses leveraging diverse patient samples from the Spanish Coalition to Unlock Research of Host Genetics on COVID-19 (SCOURGE), representing patients treated with immunomodulators (n = 849), corticoids (n = 2202), and the combined cohort of both treatments (n = 2487) who developed different outcomes. We assessed various phenotypes as indicators of treatment response, including survival at 90 days, admission to the intensive care unit (ICU), radiological affectation, and type of ventilation. Results: We identified significant polymorphisms in 16 genes from the GWAS and candidate gene studies (TLR1, TLR6, TLR10, CYP2C19, ACE2, UGT1A1, IL-1α, ZMAT3, TLR4, MIR924HG, IFNG-AS1, ABCG1, RBFOX1, ABCB11, TLR5, and ANK3) that may modulate the response to corticoid and immunomodulator therapies in COVID-19 patients. Enrichment analyses revealed overrepresentation of genes involved in the innate immune system, drug ADME, viral infection, and the programmed cell death pathways associated with the response phenotypes. Conclusions: Our study provides an initial framework for understanding the genetic determinants of treatment response in COVID-19 patients, offering insights that could inform precision medicine approaches for future epidemics.
Collapse
Affiliation(s)
| | - Natalia Cullell
- Fundació Docència i Recerca Mutua Terrassa, 08221 Terrassa, Spain;
- Hospital Universitario Mutua Terrassa, 08221 Terrassa, Spain
| | - Olalla Maroñas
- Fundación Pública Galega de Medicina Genómica (FPGMX), Centro Nacional de Genotipado (CEGEN), Servicio Gallego de Salud (SERGAS), 15706 Santiago de Compostela, Spain
- Grupo de Farmacogenómica y Descubrimiento de Medicamentos (GenDeM), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.G.-R.); (S.R.-R.)
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María José Herrero
- IIS La Fe, Plataforma de Farmacogenética, 43026 Valencia, Spain
- Departamento de Farmacología, Universidad de Valencia, 46010 Valencia, Spain
| | - Raquel Cruz
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Centro Nacional de Genotipado (CEGEN), Universidad de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Berta Almoguera
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Carmen Ayuso
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Rosario López-Rodríguez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | | | - Rocio Ortiz-Lopez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud and Hospital San Jose TecSalud, Monterrey 64718, Mexico
| | - María Barreda-Sánchez
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30120 Murcia, Spain
- Departamento de Ciencias de la Salud, Universidad Católica San Antonio de Murcia (UCAM), 30120 Murcia, Spain
| | - Marta Corton
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - David Dalmau
- Fundació Docència i Recerca Mutua Terrassa, 08221 Terrassa, Spain;
- Hospital Universitario Mutua Terrassa, 08221 Terrassa, Spain
| | - Esther Calbo
- Hospital Universitario Mutua Terrassa, 08221 Terrassa, Spain
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | | | - Beatriz Dietl
- Hospital Universitario Mutua Terrassa, 08221 Terrassa, Spain
| | - Anna Sangil
- Hospital Universitario Mutua Terrassa, 08221 Terrassa, Spain
| | - Almudena Gil-Rodriguez
- Grupo de Farmacogenómica y Descubrimiento de Medicamentos (GenDeM), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.G.-R.); (S.R.-R.)
- Grupo de Medicina Genómica, CIMUS, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Encarna Guillén-Navarro
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30120 Murcia, Spain
- Sección Genética Médica-Servicio de Pediatría, Hospital Clínico Universitario Virgen de la Arrixaca, Servicio Murciano de Salud, 30120 Murcia, Spain
- Departamento Cirugía, Pediatría, Obstetricia y Ginecología, Facultad de Medicina, Universidad de Murcia (UMU), 30120 Murcia, Spain
| | - Esther Mancebo
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Transplant Immunology and Immunodeficiencies Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | | | - Pablo Minguez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital-Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Transplant Immunology and Immunodeficiencies Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gladys G. Olivera
- IIS La Fe, Plataforma de Farmacogenética, 43026 Valencia, Spain
- Departamento de Farmacología, Universidad de Valencia, 46010 Valencia, Spain
| | - Sheila Recarey-Rama
- Grupo de Farmacogenómica y Descubrimiento de Medicamentos (GenDeM), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.G.-R.); (S.R.-R.)
- Grupo de Medicina Genómica, CIMUS, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Luis Sendra
- IIS La Fe, Plataforma de Farmacogenética, 43026 Valencia, Spain
- Departamento de Farmacología, Universidad de Valencia, 46010 Valencia, Spain
| | - Enrique G. Zucchet
- IIS La Fe, Plataforma de Farmacogenética, 43026 Valencia, Spain
- Departamento de Farmacología, Universidad de Valencia, 46010 Valencia, Spain
| | - Miguel López de Heredia
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carlos Flores
- Genomics Division, Instituto Tecnológico y de Energías Renovables, 38600 Santa Cruz de Tenerife, Spain;
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Instituto de Investigación Sanitaria de Canarias, 38010 Santa Cruz de Tenerife, Spain
- Centre for Biomedical Network Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| | - José A. Riancho
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Medicina Interna, Hospital U.M. Valdecilla, Universidad de Cantabria, IDIVAL, 39008 Santander, Spain
| | - Augusto Rojas-Martinez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud and Hospital San Jose TecSalud, Monterrey 64718, Mexico
| | - Pablo Lapunzina
- Fundación Pública Galega de Medicina Genómica (FPGMX), Centro Nacional de Genotipado (CEGEN), Servicio Gallego de Salud (SERGAS), 15706 Santiago de Compostela, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Genómica (FPGMX), Centro Nacional de Genotipado (CEGEN), Servicio Gallego de Salud (SERGAS), 15706 Santiago de Compostela, Spain
- Grupo de Medicina Genómica, CIMUS, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Grupo de Genética, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María J. Arranz
- Fundació Docència i Recerca Mutua Terrassa, 08221 Terrassa, Spain;
| | | |
Collapse
|
15
|
Saleem W, Aslam A, Tariq M, Nauwynck H. Intestinal mucus: the unsung hero in the battle against viral gastroenteritis. Gut Pathog 2025; 17:11. [PMID: 39972475 PMCID: PMC11841282 DOI: 10.1186/s13099-025-00684-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Intestinal mucus plays a crucial role in defending against enteric infections by protecting the vulnerable intestinal epithelial cells both physically and through its various constituents. Despite this, numerous gastroenteritis-causing viruses, such as rotavirus, coronavirus, adenovirus, astrovirus, calicivirus, and enterovirus, continue to pose significant threats to humans and animals. While several studies have examined the interactions between these viruses and intestinal mucus, significant gaps remain in understanding the full protective potential of intestinal mucus against these pathogens. This review aims to elucidate the protective role of intestinal mucus in viral gastroenteritis. It begins with a comprehensive literature overview of (i) intestinal mucus, (ii) enteric viruses of medical and veterinary importance, and (iii) the known interactions between various enteric viruses and intestinal mucus. Following this, a case study is presented to highlight the age-dependent blocking effect of porcine intestinal mucus against transmissible gastroenteritis virus, a porcine coronavirus. Finally, the review discusses future investigation directions to further explore the potential of intestinal mucus as a defense mechanism against viral gastroenteritis to stimulate further research in this dynamic and critical area.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium.
| | - Ateeqa Aslam
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| | - Mehlayl Tariq
- Laboratory of Biomedical Chemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, 53-114, Poland
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| |
Collapse
|
16
|
Quinn AE, Zhao L, Bell SD, Huq MH, Fang Y. Exploring Asthma as a Protective Factor in COVID-19 Outcomes. Int J Mol Sci 2025; 26:1678. [PMID: 40004141 PMCID: PMC11855143 DOI: 10.3390/ijms26041678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Asthma has long been associated with increased susceptibility to viral respiratory infections, leading to significant exacerbations and poorer clinical outcomes. Contrarily and interestingly, emerging data and research surrounding the COVID-19 pandemic have shown that patients with asthma infected with SARS-CoV-2 experienced decreased severity of disease, lower hospitalization rates, as well as decreased morbidity and mortality. Research has shown that eosinophils could enhance immune defense against viral infections, while inhaled corticosteroids can assist in controlling systematic inflammation. Moreover, reduced ACE-2 expression in individuals with asthma may restrict viral entry, and the Th2 immune response may offset the Th1 response typically observed in severe COVID-19 patients. These factors may help explain the favorable outcomes seen in asthmatic patients during the COVID-19 pandemic. This review highlights potential protective mechanisms seen in asthmatic patients, including eosinophilia, the use of inhaled corticosteroids, reduced ACE-2 expression, and a dominate Th2 immune response. Such a study will be helpful to better manage patients with asthma who have contracted COVID-19.
Collapse
Affiliation(s)
- Anthony E. Quinn
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Muhammad H. Huq
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
17
|
Khan MY, Shah AU, Duraisamy N, ElAlaoui RN, Cherkaoui M, Hemida MG. Leveraging Artificial Intelligence and Gene Expression Analysis to Identify Some Potential Bovine Coronavirus (BCoV) Receptors and Host Cell Enzymes Potentially Involved in the Viral Replication and Tissue Tropism. Int J Mol Sci 2025; 26:1328. [PMID: 39941096 PMCID: PMC11818245 DOI: 10.3390/ijms26031328] [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: 12/22/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Bovine coronavirus (BCoV) exhibits dual tissue tropism, infecting both the respiratory and enteric tracts of cattle. Viral entry into host cells requires a coordinated interaction between viral and host proteins. However, the specific cellular receptors and co-receptors facilitating BCoV entry remain poorly understood. Similarly, the roles of host proteases such as Furin, TMPRSS2, and Cathepsin-L (CTS-L), known to assist in the replication of other coronaviruses, have not been extensively explored for BCoV. This study aims to identify novel BCoV receptors and host proteases that modulate viral replication and tissue tropism. Bovine cell lines were infected with BCoV isolates from enteric and respiratory origins, and the host cell gene expression profiles post-infection were analyzed using next-generation sequencing (NGS). Differentially expressed genes encoding potential receptors and proteases were further assessed using in-silico prediction and molecular docking analysis. These analyses focused on known coronavirus receptors, including ACE2, NRP1, DPP4, APN, AXL, and CEACAM1, to identify their potential roles in BCoV infection. Validation of these findings was performed using the qRT-PCR assays targeting individual genes. We confirmed the gene expression profiles of these receptors and enzymes in some BCoV (+/-) lung tissues. Results revealed high binding affinities of 9-O-acetylated sialic acid and NRP1 to BCoV spike (S) and hemagglutinin-esterase (HE) proteins compared to ACE2, DPP4, and CEACAM1. Additionally, Furin and TMPRSS2 were predicted to interact with the BCoV-S polybasic cleavage site (RRSRR|A), suggesting their roles in S glycoprotein activation. This is the first study to explore the interactions of BCoV with multiple host receptors and proteases. Functional studies are recommended to confirm their roles in BCoV infection and replication.
Collapse
Affiliation(s)
- Mohd Yasir Khan
- Department of Computer Science, College of Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY 11201, USA; (M.Y.K.); (N.D.); (R.N.E.); (M.C.)
| | - Abid Ullah Shah
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, NY 11548, USA;
| | - Nithyadevi Duraisamy
- Department of Computer Science, College of Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY 11201, USA; (M.Y.K.); (N.D.); (R.N.E.); (M.C.)
| | - Reda Nacif ElAlaoui
- Department of Computer Science, College of Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY 11201, USA; (M.Y.K.); (N.D.); (R.N.E.); (M.C.)
| | - Mohammed Cherkaoui
- Department of Computer Science, College of Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY 11201, USA; (M.Y.K.); (N.D.); (R.N.E.); (M.C.)
| | - Maged Gomaa Hemida
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, NY 11548, USA;
| |
Collapse
|
18
|
Tseng Y. A theoretical systems chronopharmacology approach for COVID-19: Modeling circadian regulation of lung infection and potential precision therapies. CPT Pharmacometrics Syst Pharmacol 2025; 14:340-350. [PMID: 39563101 PMCID: PMC11812942 DOI: 10.1002/psp4.13277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has underscored the urgent need for innovative therapeutic approaches. Recent studies have revealed a complex interplay between the circadian clock and SARS-CoV-2 infection in lung cells, opening new avenues for targeted interventions. This systems pharmacology study investigates this intricate relationship, focusing on the circadian protein BMAL1. BMAL1 plays a dual role in viral dynamics, driving the expression of the viral entry receptor ACE2 while suppressing interferon-stimulated antiviral genes. Its critical position at the host-pathogen interface suggests potential as a therapeutic target, albeit requiring a nuanced approach to avoid disrupting essential circadian regulation. To enable precise tuning of potential interventions, we constructed a computational model integrating the lung cellular clock with viral infection components. We validated this model against literature data to establish a platform for drug administration simulation studies using the REV-ERB agonist SR9009. Our simulations of optimized SR9009 dosing reveal circadian-based strategies that potentially suppress viral infection while minimizing clock disruption. This quantitative framework offers insights into the viral-circadian interface, aiming to guide the development of chronotherapy-based antivirals. More broadly, it underscores the importance of understanding the connections between circadian timing, respiratory viral infections, and therapeutic responses for advancing precision medicine. Such approaches are vital for responding effectively to the rapid spread of coronaviruses like SARS-CoV-2.
Collapse
Affiliation(s)
- Yu‐Yao Tseng
- Department of Food Science, Nutrition, and Nutraceutical BiotechnologyShih Chien UniversityTaipeiTaiwan
| |
Collapse
|
19
|
Shaik KM, Kumar D, Srikanth P, Nandi S. SARS-CoV-2: A synergy to the Alzheimer's disease. J Neurovirol 2025; 31:16-23. [PMID: 39998800 DOI: 10.1007/s13365-025-01247-w] [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: 01/13/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
COVID-19 was a nightmare in humankind's history that challenged our advanced medical technology. All credit goes to the researchers who played a crucial role in curbing COVID-19 and proved our medical technology supremacy. However, COVID-19 has left some mysterious scars on human well-being. It is believed that COVID-19 has a significant negative impact on various cardiovascular (CVS) and central nervous system (CNS) diseases, especially in the case of CNS diseases like Alzheimer's. Surprisingly, COVID-19 affects the respiratory system, whereas Alzheimer's disease (AD) alters brain function. To explain this phenomenon, several hypotheses were proposed, but the mechanism needs to be clearly understood. Another critical thing to be concerned about is that COVID-19 will worsen pre-existing conditions and lead to the onset of AD. In the race to curb COVID-19, the invention of vaccines was speeded up, and it is necessary to fight against COVID-19. However, postvaccination follow-up is mandatory when an individual is a victim of AD. In this review article, we compiled the various dreadful effects of the COVID-19 virus on AD, the Post effects of the virus on AD, and the effect of the COVID-19 vaccination on AD. This article provides a new direction for research concerning COVID-19 and AD.
Collapse
Affiliation(s)
- Khaja Moinuddin Shaik
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Deepak Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Pirangi Srikanth
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Sukhendu Nandi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, 160 062, Punjab, India.
| |
Collapse
|
20
|
Moharram FA, Ibrahim RR, Mahgoub S, Abdel-Aziz MS, Said AM, Huang HC, Chen LY, Lai KH, Hashad N, Mady MS. Secondary metabolites of Alternaria alternate appraisal of their SARS-CoV-2 inhibitory and anti-inflammatory potentials. PLoS One 2025; 20:e0313616. [PMID: 39854441 PMCID: PMC11760621 DOI: 10.1371/journal.pone.0313616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/28/2024] [Indexed: 01/26/2025] Open
Abstract
This study identifies the secondary metabolites from Alternaria alternate and evaluates their ACE-2: Spike RBD (SARS-CoV-2) inhibitory activity confirmed via immunoblotting in human lung microvascular endothelial cells. In addition, their in vitro anti-inflammatory potential was assessed using a cell-based assay in LPS-treated RAW 264.7 macrophage cells. Two novel compounds, altenuline (1), phthalic acid bis (7'/7'' pentyloxy) isohexyl ester (2), along with 1-deoxyrubralactone (3) alternariol-5-O-methyl ether (4) and alternariol (5) were identified. Molecular docking and in vitro studies showed that compounds 2 and 4 were promising to counteract SARS-CoV-2 attachment to human ACE-2. Thus, they are considered promising natural anti-viral agents. SwissADME in silico analysis was conducted to predict the drug-like potential. Immunoblotting analysis confirmed that the tested compounds (1-4) demonstrated downregulation of ACE-2 expression in the endothelial cells from the lungs with variable degrees. Furthermore, the tested compounds (1-4) showed promising anti-inflammatory activities through TNF-α: TNFR2 inhibitory activity and their inhibitory effect on the proinflammatory cytokines (TNF-α and IL-6) in LPS-stimulated monocytes. In conclusion, our study, for the first time, provides beneficial experimental confirmation for the efficiency of the A. alternate secondary metabolites for the treatment of COVID-19 as they hinder SARS-CoV-2 infection and lower inflammatory responses initiated by SARS-CoV-2. A. alternate and its metabolites are considered in developing preventative and therapeutic tactics for COVID-19.
Collapse
Affiliation(s)
- Fatma A. Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Reham R. Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Shahenda Mahgoub
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Abdel-Aziz
- Genetic Engineering and Biotechnology Division, Microbial Chemistry Department, National Research Centre, Giza, Egypt
| | - Ahmed M. Said
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Hui-Chi Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lo-Yun Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nashwa Hashad
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
21
|
Varghese A, Liu J, Liu B, Guo W, Dong F, Patterson TA, Hong H. Analysis of Structures of SARS-CoV-2 Papain-like Protease Bound with Ligands Unveils Structural Features for Inhibiting the Enzyme. Molecules 2025; 30:491. [PMID: 39942596 PMCID: PMC11820935 DOI: 10.3390/molecules30030491] [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: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
The COVID-19 pandemic, driven by the novel coronavirus SARS-CoV-2, has drastically reshaped global health and socioeconomic landscapes. The papain-like protease (PLpro) plays a critical role in viral polyprotein cleavage and immune evasion, making it a prime target for therapeutic intervention. Numerous compounds have been identified as inhibitors of SARS-CoV-2 PLpro, with many characterized through crystallographic studies. To date, over 70 three-dimensional (3D) structures of PLpro complexed ligands have been deposited in the Protein Data Bank, offering valuable insight into ligand-binding features that could aid the discovery and development of effective COVID-19 treatments targeting PLpro. In this study, we reviewed and analyzed these 3D structures, focusing on the key residues involved in ligand interactions. Our analysis revealed that most inhibitors bind to PLpro's substrate recognition sites S3/S4 and SUb2. While these sites are highly attractive and have been extensively explored, other potential binding regions, such as SUb1 and the Zn(II) domain, are less explored and may hold untapped potential for future COVID-19 drug discovery and development. Our structural analysis provides insights into the molecular features of PLpro that could accelerate the development of novel therapeutics targeting this essential viral enzyme.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (A.V.); (J.L.); (B.L.); (W.G.); (F.D.); (T.A.P.)
| |
Collapse
|
22
|
Fan Z, Chirinos J, Yang X, Shu J, Li Y, O’Brien JM, Witschey W, Rader DJ, Gur R, Zhao B. The landscape of plasma proteomic links to human organ imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320532. [PMID: 39867388 PMCID: PMC11759249 DOI: 10.1101/2025.01.14.25320532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Plasma protein levels provide important insights into human disease, yet a comprehensive assessment of plasma proteomics across organs is lacking. Using large-scale multimodal data from the UK Biobank, we integrated plasma proteomics with organ imaging to map their phenotypic and genetic links, analyzing 2,923 proteins and 1,051 imaging traits across multiple organs. We uncovered 5,067 phenotypic protein-imaging associations, identifying both organ-specific and organ-shared proteomic relations, along with their enriched protein-protein interaction networks and biological pathways. By integrating external gene expression data, we observed that plasma proteins associated with the brain, liver, lung, pancreas, and spleen tended to be primarily produced in the corresponding organs, while proteins associated with the heart, body fat, and skeletal muscle were predominantly expressed in the liver. We also mapped key protein predictors of organ structures and showed the effective stratification capability of plasma protein-based prediction models. Furthermore, we identified 8,116 genetic-root putative causal links between proteins and imaging traits across multiple organs. Our study presents the most comprehensive pan-organ imaging proteomics map, bridging molecular and structural biology and offering a valuable resource to contextualize the complex roles of molecular pathways underlying plasma proteomics in organ systems.
Collapse
Affiliation(s)
- Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julio Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Juan Shu
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yujue Li
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Joan M. O’Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, Philadelphia, PA 19104, USA
| | - Walter Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ruben Gur
- Lifespan Brain Institute (LiBI), Children’s Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA
- Brain Behavior Laboratory, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
23
|
Zhou C, Jia R, Yang J, Liu T, Liu X, Yang L, Zhao W. The Role of Ursodeoxycholic Acid Administration During the COVID-19 Pandemic: A Questionnaire Survey. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:4601882. [PMID: 39834529 PMCID: PMC11745549 DOI: 10.1155/cjid/4601882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025]
Abstract
In December 2022, China classified COVID-19 as a category B infectious disease. This ended 2 years of close epidemiological surveillance of COVID-19. The objective of this questionnaire was to assess the infection status in the COVID-19 pandemic since December in Henan Province, China, and the prevalence of infection in people who were taking ursodeoxycholic acid (UDCA) during this period. We distributed questionnaires to patients attending the gastroenterology clinic at the First Affiliated Hospital of Henan University of Chinese Medicine. The questionnaire lasted for 3 weeks and a total of 660 were collected, of which the number of people taking UDCA was 70. This is the first investigation into the rate of infection among those taking UDCA during the time of the COVID-19 pandemic. Our results showed that the overall infection rate among those taking UDCA was 71.43% (n = 50), with a 10% (n = 7) rate of asymptomatic infections, which was significantly lower than the 85.42% (n = 504) and 6.27% (n = 37) rates among respondents who did not take. The administration of UDCA showed a trend toward reducing the rate of COVID-19 infection, but the difference was not statistically significant when compared to patients with shorter durations of medication use. While less than 30% of participants remained uninfected during the study period, indicating a potential protective effect, it is important to note that complete prevention of SARS-CoV-2 infection by UDCA was not observed.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Oncology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou 325000, Zhejiang, China
| | - Ran Jia
- Key Laboratory of Liver Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jinqiu Yang
- Key Laboratory of Liver Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Tong Liu
- Key Laboratory of Liver Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoyan Liu
- Key Laboratory of Liver Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital Affiliated to Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wenxia Zhao
- Key Laboratory of Liver Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
24
|
Pöpperl P, Stoff M, Beineke A. Alveolar Macrophages in Viral Respiratory Infections: Sentinels and Saboteurs of Lung Defense. Int J Mol Sci 2025; 26:407. [PMID: 39796262 PMCID: PMC11721917 DOI: 10.3390/ijms26010407] [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: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Respiratory viral infections continue to cause pandemic and epidemic outbreaks in humans and animals. Under steady-state conditions, alveolar macrophages (AlvMϕ) fulfill a multitude of tasks in order to maintain tissue homeostasis. Due to their anatomic localization within the deep lung, AlvMϕ are prone to detect and react to inhaled viruses and thus play a role in the early pathogenesis of several respiratory viral infections. Here, detection of viral pathogens causes diverse antiviral and proinflammatory reactions. This fact not only makes them promising research targets, but also suggests them as potential targets for therapeutic and prophylactic approaches. This review aims to give a comprehensive overview of the current knowledge about the role of AlvMϕ in respiratory viral infections of humans and animals.
Collapse
Affiliation(s)
- Pauline Pöpperl
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Melanie Stoff
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| |
Collapse
|
25
|
Uematsu T, Takai-Todaka R, Haga K, Kobayashi H, Imajima M, Kobayashi N, Katayama K, Hanaki H. Pharmacological effect of cepharanthine on SARS-CoV-2-induced disease in a Syrian hamster model. J Infect Chemother 2025; 31:102505. [PMID: 39197667 DOI: 10.1016/j.jiac.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global public health threat. Although several effective vaccines and therapeutics have been developed, continuous emergence of new variants necessitates development of drugs with different mechanisms of action. Recent studies indicate that cepharanthine, a chemical derivative purified from Stephania cepharantha, inhibits SARS-CoV-2 replication in vitro. METHODS This study examined the in vivo effects of cepharanthine using a Syrian hamster SARS-CoV-2 infection model. To evaluate the prophylactic and therapeutic effects, cepharanthine was intranasally administered before or after SARS-CoV-2 infection. Effects were assessed by monitoring body weight changes, lung pathology, lung viral load, and inflammatory response in the lungs. RESULTS Pre-infection administration of cepharanthine resulted in less weight loss, reduced virus titers, alleviated histopathological severity, and decreased lung inflammation. Furthermore, post-infection administration of cepharanthine also exhibited therapeutic effects. CONCLUSIONS This study demonstrated that both prophylactic and therapeutic administration of cepharanthine reduces the pathogenesis of COVID-19 in a Syrian hamster SARS-CoV-2 infection model. Our findings suggest that cepharanthine is a potential therapeutic agent against COVID-19.
Collapse
Affiliation(s)
- Takayuki Uematsu
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan.
| | - Reiko Takai-Todaka
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Kei Haga
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Hideyuki Kobayashi
- Tokyo New Drug Research Laboratories, Pharmaceutical Business Unit, Kowa Company, Ltd., Tokyo, Japan.
| | - Makiko Imajima
- Tokyo New Drug Research Laboratories, Pharmaceutical Business Unit, Kowa Company, Ltd., Tokyo, Japan.
| | - Noritada Kobayashi
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan.
| | - Kazuhiko Katayama
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Hideaki Hanaki
- Infection Control Research Center, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| |
Collapse
|
26
|
Benício LFMA, Nascimento ÉCM, Martins JBL. Docking heparan sulfate-based ligands as a promising inhibitor for SARS-CoV-2. J Mol Model 2024; 31:19. [PMID: 39666205 DOI: 10.1007/s00894-024-06236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
CONTEXT Heparan sulfate (HS) linear polysaccharide glycosaminoglycan compound is linked to components from the cell surface and the extracellular matrix. HS mediates SARS-CoV-2 infection through spike protein binding to cell surface receptors and is required to bind ACE2, prompting the need for electronic structure and molecular docking evaluation of this core system to exploit this attachment in developing new derivatives. Therefore, we have studied five molecules based on HS using molecular docking and electronic structure analysis. Non-covalent interaction analysis shows hydrogen bonding and van der Waals interactions in the binding to RBD-ACE2 interface and 3CLpro. SDM3 and SDM1 molecules present the lowest gap, including solvent effect under 154.6 kcal/mol, and exhibit the most reactivity behavior in this group, potentially leading to enhanced interaction in docking studies. METHODS Heparan sulfate and four derivatives were optimized using B3LYP functional with two basis sets 6-31 + G(d,p) and def2SVP. Electronic structure was used to explore the main interactions and the reactivity of these molecules, and these optimized structures were used in the molecular docking study against 3CLpro, RBD, and ACE2.
Collapse
Affiliation(s)
- Luiz F M A Benício
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Érica C M Nascimento
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - João B L Martins
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil.
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil.
| |
Collapse
|
27
|
Stich M, Magalhães VG, Bürger F, Garbade SF, Jeltsch K, Mohr K, Haddad A, Elling R, Lang P, Rabsteyn A, Jacobsen E, Bode SFN, Müller B, Kräusslich H, Hoffmann GF, Okun JG, Bartenschlager R, Binder M, Janda A, Renk H, Tönshoff B. Elevated Soluble ACE2 Activity in Children and Adults After SARS-CoV-2 Exposure Irrespective of Laboratory-Confirmed Infection. J Med Virol 2024; 96:e70098. [PMID: 39624009 PMCID: PMC11612704 DOI: 10.1002/jmv.70098] [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: 07/22/2024] [Revised: 10/21/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
The pivotal role of the cell entry receptor ACE2 for SARS-CoV-2 infection is well-established. When ACE2 is shed from cell surface into plasma as soluble ACE2 (sACE2), it can effectively neutralize SARS-CoV-2. This longitudinal prospective cohort study analyzed sACE2 activity in 1192 participants, aged 4 months to 81 years, 3 and 12 months after SARS-CoV-2 household exposure. Following SARS-CoV-2 exposure, participants exhibited significantly elevated sACE2 activity, irrespective of confirmed infection, with the highest levels observed in exposed children. Longitudinal analysis revealed a decline in sACE2 levels over time, reaching levels comparable to age- and sex-matched pre-pandemic controls. An increase in sACE2 activity was also confirmed in vitro in Calu-3 (human lung) cells within hours of SARS-CoV-2 exposure, providing a direct link between SARS-CoV-2 exposure and elevated sACE2. This study, therefore, challenges the dichotomy of categorizing SARS-CoV-2 exposed participants as infected or not infected solely on currently established diagnostic assays. It demonstrates lasting host responses independent of B- and T-cell memory and may help to keep SARS-CoV-2 infections in balance and contribute to successful virus clearance in children and adults lacking humoral and cellular immune responses following SARS-CoV-2 exposure. Trial Registration: German Registry for Clinical Studies; Identifier: D 00021521.
Collapse
Affiliation(s)
- Maximilian Stich
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, Molecular VirologyHeidelbergGermany
- German Cancer Research Center (DKFZ)Division Virus‐Associated CarcinogenesisHeidelbergGermany
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
| | - Vladimir Gonçalves Magalhães
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Friederike Bürger
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Sven F. Garbade
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Kathrin Jeltsch
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Kerstin Mohr
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Anneke Haddad
- Institute for Infection Prevention and ControlUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
- Center for Pediatrics and Adolescent MedicineUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
| | - Roland Elling
- Center for Pediatrics and Adolescent MedicineUniversity Medical Centre and Faculty of Medicine FreiburgFreiburg im BreisgauGermany
- Institute for ImmunodeficiencyMedical Center Freiburg, Germany and Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Peter Lang
- University Children's Hospital TübingenDepartment of Hematology/OncologyTübingenGermany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)Partner Site TübingenTübingenGermany
| | - Armin Rabsteyn
- University Children's Hospital TübingenDepartment of Hematology/OncologyTübingenGermany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)Partner Site TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”University of TübingenTübingenGermany
| | - Eva‐Maria Jacobsen
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Sebastian F. N. Bode
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Barbara Müller
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, VirologyHeidelbergGermany
| | - Hans‐Georg Kräusslich
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, VirologyHeidelbergGermany
| | - Georg Friedrich Hoffmann
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Jürgen G. Okun
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| | - Ralf Bartenschlager
- Heidelberg UniversityMedical Faculty Heidelberg, Department of Infectious Diseases, Molecular VirologyHeidelbergGermany
- German Cancer Research Center (DKFZ)Division Virus‐Associated CarcinogenesisHeidelbergGermany
- German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
| | - Marco Binder
- German Cancer Research Center (DKFZ)Division Virus‐Associated Carcinogenesis, Research Group “Dynamics of Early Viral Infection and the Innate Antiviral Response”HeidelbergGermany
| | - Aleš Janda
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Hanna Renk
- University Children's Hospital TübingenDepartment of Pediatric Neurology and Developmental MedicineTübingenGermany
| | - Burkhard Tönshoff
- Heidelberg UniversityMedical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Department of Pediatrics IHeidelbergGermany
| |
Collapse
|
28
|
Asrorov AM, Ayubov MS, Tu B, Shi M, Wang H, Mirzaakhmedov S, Kumar Nayak A, Abdurakhmonov IY, Huang Y. Coronavirus spike protein-based vaccines. Vaccine delivery systems. MEDICINE IN DRUG DISCOVERY 2024; 24:100198. [DOI: 10.1016/j.medidd.2024.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
|
29
|
Kumar P, Zhang X, Shaha R, Kschischo M, Dobbelstein M. Identification of antibody-resistant SARS-CoV-2 mutants via N4-Hydroxycytidine mutagenesis. Antiviral Res 2024; 231:106006. [PMID: 39293594 DOI: 10.1016/j.antiviral.2024.106006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Monoclonal antibodies targeting the Spike protein of SARS-CoV-2 are effective against COVID-19 and might mitigate future pandemics. However, their efficacy is challenged by the emergence of antibody-resistant virus variants. We developed a method to efficiently identify such resistant mutants based on selection from mutagenized virus pools. By inducing mutations with the active compound of Molnupiravir, N4-hydroxycytidine (NHC), and subsequently passaging the virus in the presence of antibodies, we identified specific Spike mutations linked to resistance. Validation of these mutations was conducted using pseudotypes and immunofluorescence analysis. From a Wuhan-like strain of SARS-CoV-2, we identified the following mutations conferring strong resistance towards the corresponding antibodies: Bamlanivimab - E484K, F490S and S494P; Sotrovimab - E340K; Cilgavimab - K444R/E and N450D. From the Omicron B.1.1.529 variant, the strongly selected mutations were: Bebtelovimab - V445A; Sotrovimab - E340K and K356M; Cilgavimab - K444R, V445A and N450D. We also identified escape mutations in the Wuhan-like Spike for the broadly neutralizing antibodies S2K146 - combined G485S and Q493R - and S2H97 - D428G, K462E and S514F. Structural analysis revealed that the selected mutations occurred at antibody-binding residues within the receptor-binding domains of the Spike protein. Most of the selected mutants largely maintained ACE2 binding and infectivity. Notably, many of the identified resistance-conferring mutations are prevalent in real-world SARS-CoV-2 variants, but some of them (G485S, D428G, and K462E) have not yet been observed in circulating strains. Our approach offers a strategy for predicting the therapeutic efficacy of antibodies against emerging virus variants.
Collapse
MESH Headings
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/drug effects
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Cytidine/genetics
- Humans
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Drug Resistance, Viral/genetics
- Mutation
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Mutagenesis
- COVID-19/virology
- COVID-19/immunology
- Antiviral Agents/pharmacology
- COVID-19 Drug Treatment
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Hydroxylamines
Collapse
Affiliation(s)
- Priya Kumar
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany
| | - Xiaoxiao Zhang
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany; Department of Informatics, Technical University of Munich, 81675, Munich, Germany
| | - Rahul Shaha
- Department of Molecular Enzymology, Göttingen Center of Molecular Biosciences (GZMB), University of Göttingen, 37077, Göttingen, Germany
| | - Maik Kschischo
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
30
|
Zaremba S, Miller AJ, Ovrom EA, Senefeld JW, Wiggins CC, Dominelli PB, Ganesh R, Hurt RT, Bartholmai BJ, Welch BT, Ripoll JG, Joyner MJ, Ramsook AH. Increased luminal area of large conducting airways in patients with COVID-19 and post-acute sequelae of COVID-19: a retrospective case-control study. J Appl Physiol (1985) 2024; 137:1168-1174. [PMID: 39298620 PMCID: PMC11573277 DOI: 10.1152/japplphysiol.00573.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is associated with enlarged luminal areas of large conducting airways. In 10-30% of patients with acute COVID-19 infection, symptoms persist for more than 4 wk (referred to as post-acute sequelae of COVID 19, or PASC), and it is unknown if airway changes are associated with this persistence. Thus, we aim to investigate whether luminal area of large conducting airways is different between patients with PASC and COVID-19 and healthy controls. In this retrospective case-control study, 75 patients with PASC (48 females) were age-, height-, and sex-matched to 75 patients with COVID-19 and 75 healthy controls. Using three-dimensional digital reconstruction from computed tomography imaging, we measured luminal areas of seven conducting airways, including trachea, right and left main bronchi, bronchus intermediate, right and left upper lobe, and left lower lobe bronchi. Kruskal-Wallis H test was used to compare measurements between the three groups, as appropriate. Airway luminal areas between COVID-19 and PASC groups were not different (all, P > 0.66). There were no group differences in airway luminal area (PASC vs. control) for trachea and right main bronchus. However, in the remaining five airways, airway luminal areas were 12-39% larger among patients with PASC than in controls (all, P < 0.05). Patients diagnosed with COVID-19 and PASC have greater airway luminal area in most large conducting airways compared with healthy controls. No differences in luminal area between patients with COVID-19 and PASC suggest persistence of changes or insufficient time for reversal of changes.NEW & NOTEWORTHY Three-dimensional reconstruction of airways has shown increased luminal area in patients with COVID-19 and post-acute sequelae of COVID-19 when compared with healthy controls. These findings suggest the role of large conducting airways in the pathogenesis of post-acute sequelae of COVID 19.
Collapse
Affiliation(s)
- Solomiia Zaremba
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Alex J Miller
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Erik A Ovrom
- Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
| | - Chad C Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Kinesiology, Michigan State University, East Lansing, Michigan, United States
| | - Paolo B Dominelli
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Ravindra Ganesh
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Ryan T Hurt
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Brian J Bartholmai
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States
| | - Brian T Welch
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States
| | - Juan G Ripoll
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Andrew H Ramsook
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
31
|
Wimalawansa SJ. Unveiling the Interplay-Vitamin D and ACE-2 Molecular Interactions in Mitigating Complications and Deaths from SARS-CoV-2. BIOLOGY 2024; 13:831. [PMID: 39452140 PMCID: PMC11504239 DOI: 10.3390/biology13100831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024]
Abstract
The interaction of the SARS-CoV-2 spike protein with membrane-bound angiotensin-converting enzyme-2 (ACE-2) receptors in epithelial cells facilitates viral entry into human cells. Despite this, ACE-2 exerts significant protective effects against coronaviruses by neutralizing viruses in circulation and mitigating inflammation. While SARS-CoV-2 reduces ACE-2 expression, vitamin D increases it, counteracting the virus's harmful effects. Vitamin D's beneficial actions are mediated through complex molecular mechanisms involving innate and adaptive immune systems. Meanwhile, vitamin D status [25(OH)D concentration] is inversely correlated with severity, complications, and mortality rates from COVID-19. This study explores mechanisms through which vitamin D inhibits SARS-CoV-2 replication, including the suppression of transcription enzymes, reduced inflammation and oxidative stress, and increased expression of neutralizing antibodies and antimicrobial peptides. Both hypovitaminosis D and SARS-CoV-2 elevate renin levels, the rate-limiting step in the renin-angiotensin-aldosterone system (RAS); it increases ACE-1 but reduces ACE-2 expression. This imbalance leads to elevated levels of the pro-inflammatory, pro-coagulatory, and vasoconstricting peptide angiotensin-II (Ang-II), leading to widespread inflammation. It also causes increased membrane permeability, allowing fluid and viruses to infiltrate soft tissues, lungs, and the vascular system. In contrast, sufficient vitamin D levels suppress renin expression, reducing RAS activity, lowering ACE-1, and increasing ACE-2 levels. ACE-2 cleaves Ang-II to generate Ang(1-7), a vasodilatory, anti-inflammatory, and anti-thrombotic peptide that mitigates oxidative stress and counteracts the harmful effects of SARS-CoV-2. Excess ACE-2 molecules spill into the bloodstream as soluble receptors, neutralizing and facilitating the destruction of the virus. These combined mechanisms reduce viral replication, load, and spread. Hence, vitamin D facilitates rapid recovery and minimizes transmission to others. Overall, vitamin D enhances the immune response and counteracts the pathological effects of SARS-CoV-2. Additionally, data suggests that widely used anti-hypertensive agents-angiotensin receptor blockers and ACE inhibitors-may lessen the adverse impacts of SARS-CoV-2, although they are less potent than vitamin D.
Collapse
|
32
|
Schermuly II, Romanet S, Patra AK, Mastrototaro L, Lemme A, Pieper R, Zentek J, Aschenbach JR. Transport of Neutral Amino Acids in the Jejunum of Pigs with Special Consideration of L-Methionine. Nutrients 2024; 16:3418. [PMID: 39408384 PMCID: PMC11478682 DOI: 10.3390/nu16193418] [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: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Methionine (Met) is a popular nutritional supplement in humans and animals. It is routinely supplemented to pigs as L-Met, DL-Met, or DL-2-hydroxy-4-(methylthio) butanoic acid (DL-HMTBA). Methods: We investigated the effect of these Met supplements on jejunal amino acid (AA) transport in male castrated Piétrain × Danbred pigs, also including a non-supplemented group. The mucosal-to-serosal flux of ten [14C]-labeled AAs (L-glutamine, glycine, L-leucine, L-lysine, L-Met, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine) was investigated at two concentrations (50 µM and 5 mM). Inhibition of apical uptake by mucosal L-Met was also measured for these AAs. The intestinal expression of apical AA transporters, angiotensin-converting enzyme II and inflammation-related genes were compared with those of a previous study. Results: Except for tryptophan and lysine at 5 mM, all AA fluxes were Na+-dependent (p ≤ 0.05), and the uptake of most AAs, except glycine and lysine, was inhibited by L-Met (p < 0.001). A correlation network existed between Na+-dependent fluxes of most AAs (except tryptophan and partly glycine). We observed the upregulation of B0AT1 (SLC6A19) (p < 0.001), the downregulation of ATB0,+ (SLC6A14) (p < 0.001) and a lower expression of CASP1, IL1β, IL8, TGFβ and TNFα in the present vs. the previous study (p < 0.001). Conclusions: The correlating AAs likely share the same Na+-dependent transporter(s). A varying effect of the Met supplement type on AA transport in the two studies might be related to a different level of supplementation or a different inflammatory status of the small intestine.
Collapse
Affiliation(s)
- Isabel I. Schermuly
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Stella Romanet
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Amlan K. Patra
- American Institute for Goat Research, Langston University, Langston, OK 73050, USA;
| | - Lucia Mastrototaro
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - Andreas Lemme
- Animal Nutrition Services, Evonik Operations GmbH, Rodenbacher Chausee 4, 63457 Hanau-Wolfgang, Germany;
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| |
Collapse
|
33
|
Buibaş FI, Cercel RA, Şerbănescu MS, Turcu AA, Dumitrescu F, Pirici D, Marinescu I, Ionovici N, Busuioc CJ, Zorilă MV, Mogoantă L. Morphopathology of the lesions induced by SARS-CoV-2 infection in the lungs. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:637-645. [PMID: 39957025 PMCID: PMC11924890 DOI: 10.47162/rjme.65.4.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection spread rapidly from China around the world, causing the worst pandemic since the beginning of the 21st century. Although the disease named coronavirus disease 2019 (COVID-19) has multiple organ symptoms, the main pathological lesions occur in the lung, causing respiratory failure, pulmonary embolism, secondary bacterial pneumonia and pulmonary fibrosis. Despite the best efforts of researchers, the pathogenesis of SARS-CoV-2-induced cellular and tissue damage in organs and systems is poorly understood. Therefore, in our study, we aimed to highlight the pulmonary lesions and their extent, which could explain the complex symptomatology presented by patients who died with acute respiratory distress syndrome (ARDS). The study was performed on a number of 36 patients diagnosed with COVID-19 who died under legally suspicious conditions, requiring autopsy within the Romanian Forensic Medicine Institutes. All patients presented a local inflammatory reaction of pneumonic type, with exudative and proliferative phenomena, with intra-alveolar and interstitial inflammatory infiltrates formed by lymphocytes, macrophages and neutrophilic granulocytes, with congested or ruptured blood vessels with intra-alveolar or interstitial hemorrhages, with multiple thrombosis, with proliferation of local fibroblasts transformed into myofibroblasts and presence of granulation tissue that remodeled the entire lung parenchyma.
Collapse
|
34
|
Huang P, Yang Z, Zhan C, Xiao X, Lian Z, Fang L, Zhong S, Xu J, Xian M, Li N, Wang X, Li J, Chen R. Alteration of the airway microbiota is associated with the progression of post-COVID-19 chronic cough in adults: a prospective study. J Genet Genomics 2024; 51:1111-1120. [PMID: 38960315 DOI: 10.1016/j.jgg.2024.06.015] [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: 05/06/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024]
Abstract
Cough is one of the most common symptoms observed in patients presenting with COVID-19, persisting for an extended duration following SARS-CoV-2 infection. We aim to describe the distribution of airway microbiota and explore its role in patients with post-COVID-19 chronic cough. A total of 57 patients experiencing persistent cough after infection were recruited during the Omicron wave of SARS-CoV-2 in China. Airway microbiota profiling is assessed in nasopharyngeal swab, nasal lavage, and induced sputum samples at 4 and 8 weeks after SARS-CoV-2 infection. Our findings reveal that bacterial families Staphylococcaceae, Corynebacteriaceae, and Enterobacteriaceae are the most prevalent in the upper airway, while Streptococcaceae, Lachnospiraceae, and Prevotellaceae emerge as the most prevalent bacterial families in the lower airway. An increase in the abundance of Staphylococcus in nasopharyngeal swab samples and of Streptococcus in induced sputum samples is observed after one month. Furthermore, the abundance of Staphylococcus identified in nasopharyngeal swab samples at the baseline period emerges as an insightful predictor for improvement in cough severity. In conclusion, dynamic alterations in the airway microbial composition may contribute to the post-COVID-19 chronic cough progression, while the compositional signatures of nasopharyngeal microbiota could reflect the improvement of this disease.
Collapse
Affiliation(s)
- Peiying Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Zhaowei Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Xiaojun Xiao
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Zexuan Lian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Liman Fang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Shuxin Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jiahan Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Mo Xian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Naijian Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Xinru Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China.
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510182, China.
| |
Collapse
|
35
|
Plastiras OE, Bouquet P, Raczkiewicz I, Belouzard S, Martin De Fourchambault E, Dhainaut J, Dacquin JP, Goffard A, Volkringer C. Virucidal activity of porphyrin-based metal-organic frameworks against highly pathogenic coronaviruses and hepatitis C virus. Mater Today Bio 2024; 28:101165. [PMID: 39221218 PMCID: PMC11364898 DOI: 10.1016/j.mtbio.2024.101165] [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: 04/29/2024] [Revised: 07/06/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
The antiviral effect of four porphyrin-based Metal-Organic Frameworks (PMOFs) with Al and Zr, namely Al-TCPP, PCN-222, PCN-223 and PCN-224 was assessed for the first time against HCoV-229E, two highly pathogenic coronaviruses (SARS-CoV-2 and MERS-CoV) and hepatitis C virus (HCV). Infection tests in vitro were done under dark or light exposure for different contact times, and it was found that 15 min of light exposure were enough to give antiviral properties to the materials, therefore inactivating HCoV-229E by 99.98 % and 99.96 % for Al-TCPP and PCN-222. Al-TCPP diminished the viral titer of SARS-CoV-2 greater than PCN-222 in the same duration of light exposure, having an effect of 99.95 % and 93.48 % respectively. Next, Al-TCPP was chosen as the best candidate possessing antiviral properties and was tested against MERS-CoV and HCV, showcasing a reduction of infectivity of 99.28 % and 98.15 % respectively for each virus. The mechanism of the antiviral activity of the four PMOFs was found to be the production of singlet oxygen 1O2 from the porphyrin ligand TCPP when exposed to visible light, by using sodium azide (NaN3) as a scavenger, that can later attack the phospholipids on the envelope of the viruses, thus preventing their entry into the cells.
Collapse
Affiliation(s)
- Orfeas-Evangelos Plastiras
- Unité de Catalyse et Chimie du Solide (UCCS), Univ. Lille, CNRS, Centrale Lille, Univ. Artois, F-59000, Lille, France
- U1019, UMR 9017, CIIL - Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université de Lille, CNRS, INSERM, CHU de Lille, 59000, Lille, France
| | - Peggy Bouquet
- Clinical Microbiology Unit, Institut Pasteur de Lille, Lille, F-59000, France
| | - Imelda Raczkiewicz
- U1019, UMR 9017, CIIL - Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université de Lille, CNRS, INSERM, CHU de Lille, 59000, Lille, France
| | - Sandrine Belouzard
- U1019, UMR 9017, CIIL - Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université de Lille, CNRS, INSERM, CHU de Lille, 59000, Lille, France
| | - Esther Martin De Fourchambault
- U1019, UMR 9017, CIIL - Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université de Lille, CNRS, INSERM, CHU de Lille, 59000, Lille, France
| | - Jeremy Dhainaut
- Unité de Catalyse et Chimie du Solide (UCCS), Univ. Lille, CNRS, Centrale Lille, Univ. Artois, F-59000, Lille, France
| | - Jean-Philippe Dacquin
- Unité de Catalyse et Chimie du Solide (UCCS), Univ. Lille, CNRS, Centrale Lille, Univ. Artois, F-59000, Lille, France
| | - Anne Goffard
- U1019, UMR 9017, CIIL - Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université de Lille, CNRS, INSERM, CHU de Lille, 59000, Lille, France
| | - Christophe Volkringer
- Unité de Catalyse et Chimie du Solide (UCCS), Univ. Lille, CNRS, Centrale Lille, Univ. Artois, F-59000, Lille, France
| |
Collapse
|
36
|
Carriero F, Rubino V, Gelzo M, Scalia G, Raia M, Ciccozzi M, Gentile I, Pinchera B, Castaldo G, Ruggiero G, Terrazzano G. Immune Profile in COVID-19: Unveiling T R3-56 Cells in SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:10465. [PMID: 39408792 PMCID: PMC11477006 DOI: 10.3390/ijms251910465] [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: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The emergence of COronaVIrus Disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presented a global health challenge since its identification in December 2019. With clinical manifestations ranging from mild respiratory symptoms to severe multi-organ dysfunction, COVID-19 continues to affect populations worldwide. The complex interactions between SARS-CoV-2 variants and the human immune system are crucial for developing effective therapies, vaccines, and preventive measures. Understanding these immune responses highlights the intricate nature of COVID-19 pathogenesis. This retrospective study analyzed, by flow cytometry approach, a cohort of patients infected with SARS-CoV-2 during the initial pandemic waves from 2020 to 2021. It focused on untreated individuals at the time of hospital admission and examined the presence of TR3-56 cells in their immune profiles during the anti-viral immune response. Our findings provide additional insights into the complex immunological dynamics of SARS-CoV-2 infection and highlight the potential role of TR3-56 cells as crucial components of the immune response. We suggest that TR3-56 cells could serve as valuable biomarkers for identifying more severe cases of COVID-19, aiding in the assessment and management of the disease.
Collapse
Affiliation(s)
- Flavia Carriero
- Dipartimento di Scienze della Salute, Università degli Studi della Basilicata, 85100 Potenza, Italy;
| | - Valentina Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Naples, Italy; (V.R.); (G.R.)
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy; (M.G.); (G.S.); (M.R.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Giulia Scalia
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy; (M.G.); (G.S.); (M.R.); (G.C.)
| | - Maddalena Raia
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy; (M.G.); (G.S.); (M.R.); (G.C.)
| | - Massimo Ciccozzi
- Unità di Epidemiologia e Statistica Medica, Università Campus Biomedico, 00128 Rome, Italy;
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (I.G.); (B.P.)
| | - Biagio Pinchera
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, 80131 Naples, Italy; (I.G.); (B.P.)
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy; (M.G.); (G.S.); (M.R.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Giuseppina Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Naples, Italy; (V.R.); (G.R.)
| | - Giuseppe Terrazzano
- Dipartimento di Scienze della Salute, Università degli Studi della Basilicata, 85100 Potenza, Italy;
| |
Collapse
|
37
|
Emam MH, Mahmoud MI, El-Guendy N, Loutfy SA. Establishment of in-house assay for screening of anti-SARS-CoV-2 protein inhibitors. AMB Express 2024; 14:104. [PMID: 39285019 PMCID: PMC11405717 DOI: 10.1186/s13568-024-01739-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/04/2024] [Indexed: 09/22/2024] Open
Abstract
Developing a potent antiviral agent to combat Coronavirus Disease-19 (COVID-19) is of critical importance as we may be at risk of the emergence of new virus strains or another pandemic recurrence. The interaction between the SARS-CoV-2 spike protein and Angiotensin Converting Enzyme 2 (ACE2) is the main protein-protein interaction (PPI) implicated in the virus entry into the host cells. Spike-ACE2 PPI represents a major target for drug intervention. We have repurposed a previously described protein-protein interaction detection method to be utilized as a drug screening assay. The assay was standardized using Chitosan nanoparticles (CNPs) as the drug and SARS-CoV-2 spike-ACE2 interaction as the PPI model. The assay was then used to screen four natural bioactive compounds: Curcumin (Cur), Gallic acid (GA), Quercetin (Q), and Silymarin (Sil), and their cytotoxicity was evaluated in vitro. Production of the spike protein and the evaluation of its activity in comparison to a standard commercial protein was part of our work as well. Here we describe a novel simple immunofluorescent screening assay to identify potential SARS-CoV-2 inhibitors that could assess the inhibitory effect of any ligand against any PPI.
Collapse
Affiliation(s)
- Merna H Emam
- Nanotechnology Research Center (NTRC), the British University in Egypt, Suez Desert Road, El-Shorouk City, P.O. Box 43, Cairo, 11837, Egypt
| | - Mohamed I Mahmoud
- Nanotechnology Research Center (NTRC), the British University in Egypt, Suez Desert Road, El-Shorouk City, P.O. Box 43, Cairo, 11837, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, 11829, Cairo, Egypt
| | - Nadia El-Guendy
- Medical biochemistry and Molecular biology unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University, Fom El-Khalig 11796, Cairo, Egypt
| | - Samah A Loutfy
- Nanotechnology Research Center (NTRC), the British University in Egypt, Suez Desert Road, El-Shorouk City, P.O. Box 43, Cairo, 11837, Egypt.
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University, Fom El-Khalig 11796, Cairo, Egypt.
| |
Collapse
|
38
|
Li J, Kong X, Liu T, Xian M, Wei J. The Role of ACE2 in Neurological Disorders: From Underlying Mechanisms to the Neurological Impact of COVID-19. Int J Mol Sci 2024; 25:9960. [PMID: 39337446 PMCID: PMC11431863 DOI: 10.3390/ijms25189960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has become a hot topic in neuroscience research in recent years, especially in the context of the global COVID-19 pandemic, where its role in neurological diseases has received widespread attention. ACE2, as a multifunctional metalloprotease, not only plays a critical role in the cardiovascular system but also plays an important role in the protection, development, and inflammation regulation of the nervous system. The COVID-19 pandemic further highlights the importance of ACE2 in the nervous system. SARS-CoV-2 enters host cells by binding to ACE2, which may directly or indirectly affect the nervous system, leading to a range of neurological symptoms. This review aims to explore the function of ACE2 in the nervous system as well as its potential impact and therapeutic potential in various neurological diseases, providing a new perspective for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng 475004, China
| | - Xiangrui Kong
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng 475004, China
| |
Collapse
|
39
|
Wang YH, Lin CW, Huang CW. Polyunsaturated Fatty Acids as Potential Treatments for COVID-19-Induced Anosmia. Biomedicines 2024; 12:2085. [PMID: 39335598 PMCID: PMC11428228 DOI: 10.3390/biomedicines12092085] [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: 07/28/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Some individuals with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) experience anosmia, or loss of smell. Although the prevalence of anosmia has decreased with the emergence of the Omicron variant, it remains a significant concern. This review examines the potential role of polyunsaturated fatty acids (PUFAs), particularly omega-3 PUFAs, in treating COVID-19-induced anosmia by focusing on the underlying mechanisms of the condition. Omega-3 PUFAs are known for their anti-inflammatory, neuroprotective, and neurotransmission-enhancing properties, which could potentially aid in olfactory recovery. However, study findings are inconsistent. For instance, a placebo-controlled randomized clinical trial found no significant effect of omega-3 PUFA supplementation on olfactory recovery in patients with COVID-19-induced anosmia. These mixed results highlight the limitations of existing research, including small sample sizes, lack of placebo controls, short follow-up periods, and combined treatments. Therefore, more rigorous, large-scale studies are urgently needed to definitively assess the therapeutic potential of omega-3 PUFAs for olfactory dysfunction. Further research is also crucial to explore the broader role of PUFAs in managing viral infections and promoting sensory recovery.
Collapse
Affiliation(s)
- Yu-Han Wang
- Department of Education, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chung-Wei Lin
- Department of Education, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chiung-Wei Huang
- Department of Physiology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
40
|
Drzymała A. The Functions of SARS-CoV-2 Receptors in Diabetes-Related Severe COVID-19. Int J Mol Sci 2024; 25:9635. [PMID: 39273582 PMCID: PMC11394807 DOI: 10.3390/ijms25179635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is considered a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor of high importance, but due to its non-ubiquitous expression, studies of other proteins that may participate in virus internalisation have been undertaken. To date, many alternative receptors have been discovered. Their functioning may provide an explanation for some of the events observed in severe COVID-19 that cannot be directly explained by the model in which ACE2 constitutes the central point of infection. Diabetes mellitus type 2 (T2D) can induce severe COVID-19 development. Although many mechanisms associated with ACE2 can lead to increased SARS-CoV-2 virulence in diabetes, proteins such as basigin (CD147), glucose-regulated protein 78 kDa (GRP78), cluster of differentiation 4 (CD4), transferrin receptor (TfR), integrins α5β1/αvβ3, or ACE2 co-receptors neuropilin 2 (NRP2), vimentin, and even syalilated gangliosides may also be responsible for worsening the COVID-19 course. On the other hand, some others may play protective roles. Understanding how diabetes-associated mechanisms can induce severe COVID-19 via modification of virus receptor functioning needs further extensive studies.
Collapse
Affiliation(s)
- Adam Drzymała
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052 Opole, Poland
| |
Collapse
|
41
|
Dai P, Ma C, Jiang T, Shi J, Liu S, Zheng M, Zhou Y, Li X, Liu Y, Chen H. CD147 mediates S protein pseudovirus of SARS-CoV-2 infection and its induction of spermatogonia apoptosis. Endocrine 2024; 85:1435-1445. [PMID: 38824220 DOI: 10.1007/s12020-024-03891-4] [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: 02/23/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024]
Abstract
Male cases diagnosed COVID-19 with more complications and higher mortality compared with females, and the overall consequences of male sex hormones and semen parameters deterioration were observed in COVID-19 patients, whereas the involvement and mechanism for spermatogenic cell remains unclear. The study was aimed to investigate the infection mode of S protein (D614G) pseudovirus (pseu-S-D614G) to spermatogenic cells, as well as the influence on cell growth. Both mouse spermatogonia (GC-1 cell, immortalized spermatogonia) and spermatocyte (GC-2 cell, immortalized spermatocytes) were used to detect the infection of pseu-S-D614G of SARS-CoV-2, and further explored the effect of SARS-CoV-2-spike protein (S-protein) and SARS-CoV-2-spike protein (omicron) (O-protein) on GC-1 cell apoptosis and proliferation. The data showed that the pseu-S-D614G invaded into GC-1 cells through either human ACE2 (hACE2) or human CD147 (hCD147), whereas GC-2 cells were insensitive to viral infection. In addition, the apoptosis and proliferation suppression inflicted by S-protein and O-protein on GC-1 cells was through Bax-Caspase3 signaling rather than arresting cell cycle progression. These findings suggest that CD147, apart from ACE2, may be a potential receptor for SARS-CoV-2 infection in testicular tissues, and that the apoptotic effect was induced in spermatogonia cells by S-protein or O-protein, eventually resulted in the damage to male fertility.
Collapse
Affiliation(s)
- Pengyuan Dai
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Chaoye Ma
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Ting Jiang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Jianwu Shi
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Sha Liu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Meihua Zheng
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Yiwen Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Xiaofeng Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Lianhua Road No. 1120, Futian District, Shenzhen, Guangdong Province, PR China
| | - Yang Liu
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China.
| | - Hao Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China.
- Guangzhou Women and Children's Medical Center, GMU-GIBH Joint school of Life Science, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Deora N, Venkatraman K. Potential use of plant-based therapeutics for the management of SARS-COV2 infection in diabetes mellitus – a review. J Herb Med 2024; 47:100923. [DOI: 10.1016/j.hermed.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
43
|
Bolsinger MM, Drobny A, Wilfling S, Reischl S, Krach F, Moritz R, Balta D, Hehr U, Sock E, Bleibaum F, Hanses F, Winner B, Huarcaya SP, Arnold P, Zunke F. SARS-CoV-2 Spike Protein Induces Time-Dependent CTSL Upregulation in HeLa Cells and Alveolarspheres. J Cell Biochem 2024; 125:e30627. [PMID: 38971996 DOI: 10.1002/jcb.30627] [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: 03/27/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024]
Abstract
Autophagy and lysosomal pathways are involved in the cell entry of SARS-CoV-2 virus. To infect the host cell, the spike protein of SARS-CoV-2 binds to the cell surface receptor angiotensin-converting enzyme 2 (ACE2). To allow the fusion of the viral envelope with the host cell membrane, the spike protein has to be cleaved. One possible mechanism is the endocytosis of the SARS-CoV-2-ACE2 complex and subsequent cleavage of the spike protein, mainly by the lysosomal protease cathepsin L. However, detailed molecular and dynamic insights into the role of cathepsin L in viral cell entry remain elusive. To address this, HeLa cells and iPSC-derived alveolarspheres were treated with recombinant SARS-CoV-2 spike protein, and the changes in mRNA and protein levels of cathepsins L, B, and D were monitored. Additionally, we studied the effect of cathepsin L deficiency on spike protein internalization and investigated the influence of the spike protein on cathepsin L promoters in vitro. Furthermore, we analyzed variants in the genes coding for cathepsin L, B, D, and ACE2 possibly associated with disease progression using data from Regeneron's COVID Results Browser and our own cohort of 173 patients with COVID-19, exhibiting a variant of ACE2 showing significant association with COVID-19 disease progression. Our in vitro studies revealed a significant increase in cathepsin L mRNA and protein levels following exposure to the SARS-CoV-2 spike protein in HeLa cells, accompanied by elevated mRNA levels of cathepsin B and D in alveolarspheres. Moreover, an increase in cathepsin L promoter activity was detected in vitro upon spike protein treatment. Notably, the knockout of cathepsin L resulted in reduced internalization of the spike protein. The study highlights the importance of cathepsin L and lysosomal proteases in the SARS-CoV-2 spike protein internalization and suggests the potential of lysosomal proteases as possible therapeutic targets against COVID-19 and other viral infections.
Collapse
Affiliation(s)
- Magdalena M Bolsinger
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Stephanie Reischl
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Krach
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raul Moritz
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Denise Balta
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ute Hehr
- Center for Human Genetics Regensburg, Regensburg, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Bleibaum
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Frank Hanses
- Emergency Department, University Hospital Regensburg, Regensburg, Germany
- Department for Infection Control and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susy Prieto Huarcaya
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Philipp Arnold
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
44
|
Qin Z, Liu Y, Jia X, Zhou J, Li H, Wang X, Zhang S, Chang H, Wang G. One-step synthesized multisize AuAg alloy nanoparticles with high SERS sensitivity in directly detecting SARS-CoV-2 spike protein. Anal Chim Acta 2024; 1317:342919. [PMID: 39030015 DOI: 10.1016/j.aca.2024.342919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/21/2024]
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in widespread disease transmission, challenging the stability of global healthcare systems. Surface-enhanced Raman scattering (SERS) as an easy operation, fast, and low-cost technology illustrates a good potential in detecting SARS-CoV-2. In the study, one-step fabrication of gold-silver alloy nanoparticles (AuAgNPs) with adjustable metal proportions and diameters is employed as SERS substrates. The angiotensin-converting enzyme 2 (ACE2) functionalized AuAgNPs are applied as sensor surfaces to detect SARS-CoV-2 S protein. By optimizing the SERS substrates, ACE2/Au35Ag65NPs illustrate higher performance in detecting the SARS-CoV-2 S protein with a limit of detection (LOD) of 10 fg/mL in both phosphate-buffered saline (PBS) and pharyngeal swabs solution (PSS). It also provides excellent reproducibility with a relative standard deviation (RSD) of 7.7 % and 7.9 %, respectively. This easily preparable and highly reproducible SERS substrate has good potential in the practical application of detecting SARS-CoV-2.
Collapse
Affiliation(s)
- Zhenle Qin
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Yansheng Liu
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China.
| | - Xiaobo Jia
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Jin Zhou
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Hongli Li
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Xiaohong Wang
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Shaohui Zhang
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China
| | - Haixin Chang
- Quantum-Nano Matter and Device Lab, State Key Laboratory of Material Processing and Die and Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology Wuhan, 430074, Hubei, China
| | - Guofu Wang
- School of Electronic Engineering, Guangxi University of Science and Technology, No.2, Wenchang Road, Liuzhou City, 545006, Guangxi, China.
| |
Collapse
|
45
|
Febrianti IK, Putra AE, Raveinal R, Elliyanti A. Transcriptomic analysis of profibrinolytic and fibrinolytic inhibitor genes in COVID-19 patients. NARRA J 2024; 4:e843. [PMID: 39280271 PMCID: PMC11391964 DOI: 10.52225/narra.v4i2.843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/25/2024] [Indexed: 09/18/2024]
Abstract
The immunopathogenesis of COVID-19 infection is initiated by the entry of the SARS-CoV-2 virus into the human body through droplets, entering the lungs and binding to the ACE-2 receptor. Activated macrophages stimulate an immune and inflammatory response, leading to the activation of the coagulation cascade, including profibrinolytic and fibrinolytic inhibitor processes. One of the proteins involved in profibrinolytic is encoded by the PLAUR gene, while fibrinolytic inhibitor proteins are encoded by the A2M and SERPINE1 genes. This research aims to assess the transcriptomic analysis of genetic expression data of profibrinolytic genes, fibrinolytic inhibitor genes and their correlation with serum D-dimer levels, which describe the clinical condition of coagulation in COVID-19 patients. This cross-sectional study included 25 patients each for mild and moderate-to-severe COVID-19 at Dr. M. Djamil Padang General Hospital, Padang, Indonesia. Inter-group gene expression comparisons will be analyzed using log2 folds change, and bivariate tests will be analyzed using correlation. The results show that the PLAUR gene has higher expression in moderate-to-severe compared to mild cases. Similarly, the SERPINE1 and A2M genes expressions are higher in moderate-to-severe compared to mild cases. Furthermore, there is a significant correlation between serum D-dimer levels and profibrinolytic factor (PLAUR gene) expression in COVID-19 patients. The correlation between serum D-dimer levels with fibrinolytic inhibitor factor (SERPINE1 and A2M genes) expression was found. These conclude that there is a significant difference in the expression of the profibrinolytic and fibrinolytic inhibitor genes between mild and moderate-to-severe cases in COVID-19, demonstrating COVID-19 infection affects coagulation activities.
Collapse
Affiliation(s)
- Ika K Febrianti
- Doctoral Program of Biomedical, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
- Department of Internal Medicine, Regional Public Hospital District of Agam, Lubuk Basung, Indonesia
| | - Andani E Putra
- Department of Microbiology, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
- Dr. M. Djamil General Hospital, Padang, Indonesia
| | - Raveinal Raveinal
- Dr. M. Djamil General Hospital, Padang, Indonesia
- Department of Internal Medicine, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Aisyah Elliyanti
- Dr. M. Djamil General Hospital, Padang, Indonesia
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| |
Collapse
|
46
|
Li X, Zhao X, Yu X, Zhao J, Fang X. Construction of a multi-tissue compound-target interaction network of Qingfei Paidu decoction in COVID-19 treatment based on deep learning and transcriptomic analysis. J Bioinform Comput Biol 2024; 22:2450016. [PMID: 39036847 DOI: 10.1142/s0219720024500161] [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: 07/23/2024]
Abstract
The Qingfei Paidu decoction (QFPDD) is a widely acclaimed therapeutic formula employed nationwide for the clinical management of coronavirus disease 2019 (COVID-19). QFPDD exerts a synergistic therapeutic effect, characterized by its multi-component, multi-target, and multi-pathway action. However, the intricate interactions among the ingredients and targets within QFPDD and their systematic effects in multiple tissues remain undetermined. To address this, we qualitatively characterized the chemical components of QFPDD. We integrated multi-tissue transcriptomic analysis with GraphDTA, a deep learning model, to screen for potential compound-target interactions of QFPDD in multiple tissues. We predicted 13 key active compounds, 127 potential targets and 27 pathways associated with QFPDD across six different tissues. Notably, oleanolic acid-AXL exhibited leading affinity in the heart, blood, and liver. Molecular docking and molecular dynamics simulation confirmed their strong binding affinity. The robust interaction between oleanolic acid and the AXL receptor suggests that AXL is a promising target for developing clinical intervention strategies. Through the construction of a multi-tissue compound-target interaction network, our study further elucidated the mechanisms through which QFPDD effectively combats COVID-19 in multiple tissues. Our work also establishes a framework for future investigations into the systemic effects of other Traditional Chinese Medicine (TCM) formulas in disease treatment.
Collapse
Affiliation(s)
- Xia Li
- Third Clinical College, Shanxi Provincial Integrated TCM and WM Hospital, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, P. R. China
| | - Xuetong Zhao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xinjian Yu
- Quantitative and Computational Biosciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianping Zhao
- Third Clinical College, Shanxi Provincial Integrated TCM and WM Hospital, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, P. R. China
| | - Xiangdong Fang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
47
|
Loo C, Treacy MG, Toerien L, Msellati A, Catanzano T. Emergency Presentations of Coronavirus Disease (COVID-19): A Review of the Literature and Radiologic Perspective. Semin Ultrasound CT MR 2024; 45:332-338. [PMID: 38996944 DOI: 10.1053/j.sult.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the debilitating global pandemic known as Coronavirus disease (COVID-19). In this paper, we highlight the widespread manifestations and complications across disease systems. In addition, we present their relevant imaging findings to inform appropriate investigations and management in patients presenting to the Emergency Department with COVID-19 and its respective sequalae. Of note, we outline considerations for diagnosis of long COVID, an important medium to long term sequalae in patients with previous COVID-19 infections.
Collapse
Affiliation(s)
- Caitlyn Loo
- School of Medicine, University College Dublin, Belfield, Ireland; Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Molly Godson Treacy
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Lara Toerien
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | | | - Tara Catanzano
- Department of Radiology, Baystate Health, Springfield, MA.
| |
Collapse
|
48
|
Yu Q, Zhou X, Kapini R, Arsecularatne A, Song W, Li C, Liu Y, Ren J, Münch G, Liu J, Chang D. Cytokine Storm in COVID-19: Insight into Pathological Mechanisms and Therapeutic Benefits of Chinese Herbal Medicines. MEDICINES (BASEL, SWITZERLAND) 2024; 11:14. [PMID: 39051370 PMCID: PMC11270433 DOI: 10.3390/medicines11070014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/20/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Cytokine storm (CS) is the main driver of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) in severe coronavirus disease-19 (COVID-19). The pathological mechanisms of CS are quite complex and involve multiple critical molecular targets that turn self-limited and mild COVID-19 into a severe and life-threatening concern. At present, vaccines are strongly recommended as safe and effective treatments for preventing serious illness or death from COVID-19. However, effective treatment options are still lacking for people who are at the most risk or hospitalized with severe disease. Chinese herbal medicines have been shown to improve the clinical outcomes of mild to severe COVID-19 as an adjunct therapy, particular preventing the development of mild to severe ARDS. This review illustrates in detail the pathogenesis of CS-involved ARDS and its associated key molecular targets, cytokines and signalling pathways. The therapeutic targets were identified particularly in relation to the turning points of the development of COVID-19, from mild symptoms to severe ARDS. Preclinical and clinical studies were reviewed for the effects of Chinese herbal medicines together with conventional therapies in reducing ARDS symptoms and addressing critical therapeutic targets associated with CS. Multiple herbal formulations, herbal extracts and single bioactive phytochemicals with or without conventional therapies demonstrated strong anti-CS effects through multiple mechanisms. However, evidence from larger, well-designed clinical trials is lacking and their detailed mechanisms of action are yet to be well elucidated. More research is warranted to further evaluate the therapeutic value of Chinese herbal medicine for CS in COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Qingyuan Yu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
- Xiyuan Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Rotina Kapini
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Anthony Arsecularatne
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Wenting Song
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Chunguang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Gerald Münch
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| |
Collapse
|
49
|
Wijerathne SVT, Pandit R, Ipinmoroti AO, Crenshaw BJ, Matthews QL. Feline coronavirus influences the biogenesis and composition of extracellular vesicles derived from CRFK cells. Front Vet Sci 2024; 11:1388438. [PMID: 39091390 PMCID: PMC11292801 DOI: 10.3389/fvets.2024.1388438] [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: 02/19/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Coronavirus (CoV) has become a public health crisis that causes numerous illnesses in humans and certain animals. Studies have identified the small, lipid-bound structures called extracellular vesicles (EVs) as the mechanism through which viruses can enter host cells, spread, and evade the host's immune defenses. EVs are able to package and carry numerous viral compounds, including proteins, genetic substances, lipids, and receptor proteins. We proposed that the coronavirus could alter EV production and content, as well as influence EV biogenesis and composition in host cells. Methods In the current research, Crandell-Rees feline kidney (CRFK) cells were infected with feline coronavirus (FCoV) in an exosome-free media at a multiplicity of infection (MOI) of 2,500 infectious units (IFU) at 48 h and 72 h time points. Cell viability was analyzed and found to be significantly decreased by 9% (48 h) and 15% (72 h) due to FCoV infection. EVs were isolated by ultracentrifugation, and the surface morphology of isolated EVs was analyzed via Scanning Electron Microscope (SEM). Results NanoSight particle tracking analysis (NTA) confirmed that the mean particle sizes of control EVs were 131.9 nm and 126.6 nm, while FCoV infected-derived EVs were 143.4 nm and 120.9 nm at 48 and 72 h, respectively. Total DNA, RNA, and protein levels were determined in isolated EVs at both incubation time points; however, total protein was significantly increased at 48 h. Expression of specific protein markers such as TMPRSS2, ACE2, Alix, TSG101, CDs (29, 47, 63), TLRs (3, 6, 7), TNF-α, and others were altered in infection-derived EVs when compared to control-derived EVs after FCoV infection. Discussion Our findings suggested that FCoV infection could alter the EV production and composition in host cells, which affects the infection progression and disease evolution. One purpose of studying EVs in various animal coronaviruses that are in close contact with humans is to provide significant information about disease development, transmission, and adaptation. Hence, this study suggests that EVs could provide diagnostic and therapeutic applications in animal CoVs, and such understanding could provide information to prevent future coronavirus outbreaks.
Collapse
Affiliation(s)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL, United States
| | | | | | - Qiana L. Matthews
- Microbiology Program, Alabama State University, Montgomery, AL, United States
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
50
|
Murdocca M, Romeo I, Citro G, Latini A, Centofanti F, Bugatti A, Caccuri F, Caruso A, Ortuso F, Alcaro S, Sangiuolo F, Novelli G. A Dynamic and Effective Peptide-Based Strategy for Promptly Addressing Emerging SARS-CoV-2 Variants of Concern. Pharmaceuticals (Basel) 2024; 17:891. [PMID: 39065742 PMCID: PMC11279616 DOI: 10.3390/ph17070891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Genomic surveillance based on sequencing the entire genetic code of SARS-CoV-2 involves monitoring and studying genetic changes and variations in disease-causing organisms such as viruses and bacteria. By tracing the virus, it is possible to prevent epidemic spread in the community, ensuring a 'precision public health' strategy. A peptide-based design was applied to provide an efficacious strategy that is able to counteract any emerging viral variant of concern dynamically and promptly to affect the outcomes of a pandemic at an early stage while waiting for the production of the anti-variant-specific vaccine, which require longer times. The inhibition of the interaction between the receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and one of the cellular receptors (DPP4) that its receptors routinely bind to infect human cells is an intriguing therapeutic approach to prevent the virus from entering human cells. Among the other modalities developed for this purpose, peptides surely offer unique advantages, including ease of synthesis, serum stability, low immunogenicity and toxicity, and small production and distribution chain costs. Here, we obtained a potent new inhibitor based on the rearrangement of a previously identified peptide that has been rationally designed on a cell dipeptidyl peptidase 4 (DPP4) sequence, a ubiquitous membrane protein known to bind the RBD-SPIKE domain of the virus. This novel peptide (named DPP4-derived), conceived as an endogenous "drug", is capable of targeting the latest tested variants with a high affinity, reducing the VSV* DG-Fluc pseudovirus Omicron's infection capacity by up to 14%, as revealed by in vitro testing in human Calu-3 cells. Surface plasmon resonance (SPR) confirmed the binding affinity of the new DPP4-derived peptide with Omicron variant RBD.
Collapse
Affiliation(s)
- Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
| | - Isabella Romeo
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (I.R.); (F.O.); (S.A.)
- Net4Science Srl Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Gennaro Citro
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
| | - Andrea Latini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
| | - Federica Centofanti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
| | - Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.C.); (A.C.)
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.C.); (A.C.)
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.C.); (A.C.)
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (I.R.); (F.O.); (S.A.)
- Net4Science Srl Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (I.R.); (F.O.); (S.A.)
- Net4Science Srl Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (G.C.); (A.L.); (F.C.); (G.N.)
- IRCCS Neuromed Mediterranean Neurological Institute, 86077 Pozzilli, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|