1
|
Delic D, Klein T, Wohnhaas CT, Feng H, Lin X, Zhang JR, Wu D. Dipeptidyl peptidase-4 inhibitor linagliptin reduces inflammatory response, ameliorates tissue edema formation, and improves survival in severe sepsis. Biomed Pharmacother 2025; 182:117778. [PMID: 39724680 DOI: 10.1016/j.biopha.2024.117778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Excessive inflammation in sepsis causes microvascular dysfunction associated with organ dysfunction and high mortality. The present studies aimed to examine the therapeutic potential of linagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor in a clinically relevant polymicrobial sepsis model in mice. METHODS Sepsis was induced by cecal ligation and puncture (CLP). Mice were grouped into: Sham control+vehicle; Group 2: CLP+vehicle; Group 3: CLP+dexamethasone (10 mg/kg, s.c.) given 6 h after CLP; Group 4: CLP+linagliptin (1 mg/kg, s.c.) given 6 h after CLP. The experiment was terminated 24 hours after CLP in two experimental sets. Seven-day survival following CLP was determined in a third experimental set. RESULTS Treatment with linagliptin inhibited DPP-4 activity, increased the levels of active forms of endogenous gastric inhibitory polypeptide and glucagon-like peptide-1, without affecting the blood glucose levels in CLP mice. Compared to vehicle treatment, administration of linagliptin reduced sepsis-induced tissue hyper permeability as evidenced by a reduction in vascular Evans blue leakage, prevented edema formation in the lung, heart, liver and kidney. Furthermore, linagliptin or dexamethasone reduced sepsis-induced proinflammatory cytokine and chemokine production, such as IL-1β, IL-2, IL-10, IL-23, IL-27, VCAM-1, eotaxin, MDC, MCSF1, GCP-2, and NGAL. Importantly, administration of linagliptin improved the 7-day survival rate following CLP in mice. RNA sequencing in lung and heart revealed that linagliptin attenuated key inflammatory pathways including TNF alpha (via NFκB) and IL6/JAK/STAT3 signaling and activated interferon signaling in the heart. CONCLUSIONS Linagliptin treatment can attenuate the inflammatory response, protect against severe sepsis-induced vascular hyperpermeability, reduce multiorgan injury, and most importantly, improve the survival.
Collapse
Affiliation(s)
- Denis Delic
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Huiying Feng
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Xinchun Lin
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Jin-Rui Zhang
- First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Dongmei Wu
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA.
| |
Collapse
|
2
|
Arppo A, Barker H, Parkkila S. Bioinformatic characterization of ENPEP, the gene encoding a potential cofactor for SARS-CoV-2 infection. PLoS One 2024; 19:e0307731. [PMID: 39661628 PMCID: PMC11633960 DOI: 10.1371/journal.pone.0307731] [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: 07/10/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Research on SARS-CoV-2, the viral pathogen that causes COVID-19, has identified angiotensin converting enzyme 2 (ACE2) as the primary viral receptor. Several genes that encode viral cofactors, such as TMPRSS2, NRP1, CTSL, and possibly KIM1, have since been discovered. Glutamyl aminopeptidase (APA), encoded by the gene ENPEP, is another cofactor candidate due to similarities in its biological role and high correlation with ACE2 and other human coronavirus receptors, such as aminopeptidase N (APN) and dipeptidyl peptidase 4 (DPP4). Recent studies have proposed a role for ENPEP as a viral receptor in humans, and ENPEP and ACE2 are both closely involved in the renin-angiotensin-aldosterone system proposed to play an important role in SARS-CoV-2 pathophysiology. We performed bioinformatic analyses using publicly available bulk (>17,000 samples from 49 distinct tissues) and single-cell (>2.5 million cells) RNA-Seq gene expression datasets to evaluate the expression and function of the ENPEP gene. We also investigated age- and sex-related changes in ENPEP expression. Overall, expression of ENPEP was highest in the small intestine enterocyte brush border and the kidney cortex. ENPEP is widely expressed in a subset of vascular smooth muscle cells (likely pericytes) in systemic vasculature, the heart, and the brain. ENPEP is expressed at low levels in the lower respiratory epithelium. In the lung, ENPEP is most highly expressed in para-alveolar fibroblasts. Single-cell data revealed ENPEP expression in a substantial fraction of ependymal cells, a finding not reported before in humans. Age increases ENPEP expression in skeletal muscle and the prostate, while decreasing it in the heart and aorta. Angiogenesis was found to be a central biological function associated with the ENPEP gene. Tissue-specific roles, such as protein digestion and fat metabolism, were also identified in the intestine. In the liver, the gene is linked to the complement system, a connection that has not yet been thoroughly investigated. Expression of ENPEP and ACE2 is strongly correlated in the small intestine and renal cortex. Both overall and in blood vessels, ENPEP and ACE2 have a stronger correlation than many other genes associated with SARS-CoV-2, such as TMPRSS2, CTSL, and NRP1. Possible interaction between glutamyl aminopeptidase and SARS-CoV-2 should be investigated experimentally.
Collapse
Affiliation(s)
- Antti Arppo
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Harlan Barker
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories PLC, Tampere University Hospital, Tampere, Finland
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories PLC, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
3
|
Che Mohd Nassir CMN, Che Ramli MD, Jaffer U, Abdul Hamid H, Mehat MZ, Mohamad Ghazali M, Kottakal Cheriya EN. Neurological Sequelae of Post-COVID-19 Fatigue: A Narrative Review of Dipeptidyl Peptidase IV-Mediated Cerebrovascular Complications. Curr Issues Mol Biol 2024; 46:13565-13582. [PMID: 39727939 PMCID: PMC11727395 DOI: 10.3390/cimb46120811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/09/2024] [Accepted: 11/17/2024] [Indexed: 12/28/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic affecting millions of people's lives, which has led to 'post-COVID-19 fatigue'. Alarmingly, severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) not only infects the lungs but also influences the heart and brain. Endothelial cell dysfunction and hypercoagulation, which we know occur with this infection, lead to thrombo-inflammation that can manifest as many myriad cardio-cerebrovascular disorders, such as brain fog, fatigue, cognitive dysfunction, etc. Additionally, SARS-CoV-2 has been associated with oxidative stress, protein aggregation, cytokine storm, and mitochondrial dysfunction in neurodegenerative diseases. Accordingly, the identification of molecular targets involved in these actions could provide strategies for preventing and treating this disease. In particular, the very common enzyme dipeptidyl peptidase IV (DPPIV) has recently been identified as a candidate co-receptor for the cell entry of the SARS-CoV-2 virus with its involvement in infection. In addition, DPPIV has been reported as a co-receptor for some viruses such as Middle East respiratory syndrome-coronavirus (MERS-CoV). It mediates immunologic reactions and diseases such as type 2 diabetes mellitus, obesity, and hypertension, which have been considered the prime risk factors for stroke among other types of cardio-cerebrovascular diseases. Unlike angiotensin-converting enzyme 2 (ACE2), DPPIV has been implicated in aggravating the course of infection due to its disruptive effect on inflammatory signaling networks and the neuro-glia-vascular unit. Regarding the neurological, physiological, and molecular grounds governing post-COVID-19 fatigue, this review focuses on DPPIV as one of such reasons that progressively establishes cerebrovascular grievances following SARS-CoV infection.
Collapse
Affiliation(s)
- Che Mohd Nasril Che Mohd Nassir
- Department of Anatomy and Physiology, Faculty of Medicine, School of Basic Medical Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Terengganu, Malaysia;
| | - Muhammad Danial Che Ramli
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam 40150, Selangor, Malaysia
| | - Usman Jaffer
- Kulliyyah of Islamic Revealed Knowledge and Human Sciences, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia;
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (H.A.H.); (M.Z.M.)
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (H.A.H.); (M.Z.M.)
| | - Mazira Mohamad Ghazali
- Department of Anatomy and Physiology, Faculty of Medicine, School of Basic Medical Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Terengganu, Malaysia;
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| | | |
Collapse
|
4
|
Wankhade N, Sharma A, Wani MA, Banerjee A, Garg P. Predictive Modeling and Drug Repurposing for Type-II Diabetes. ACS Med Chem Lett 2024; 15:1907-1917. [PMID: 39563823 PMCID: PMC11571088 DOI: 10.1021/acsmedchemlett.4c00358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024] Open
Abstract
Diabetes mellitus (DM) is a global health concern, and dipeptidyl peptidase-4 (DPP-4) is a key therapeutic target. The study used three machine learning and deep learning models to predict potential DPP-4 inhibitors using a curated data set of 6,750 compounds. The models included support vector machine (SVM), random forest (RF), naive Bayes (NB), and multitask deep neural network (MTDNN). The MTDNN model demonstrated strong predictive performance, achieving 98.62% train accuracy and 98.42% test accuracy for predicting DPP-4 inhibitors and a correlation coefficient of 0.979 for training and 0.977 for the test data set, with low training and test errors while predicting corresponding IC50 values. The MTDNN model predicted potential inhibitors using an external data set of FDA-approved drugs, identifying 100 compounds. Among these, five compounds stood out with promising molecular docking and dynamic profiles, suggesting their potential as repurposed drugs for targeting DPP-4 and offering hope for the future of diabetes treatment.
Collapse
Affiliation(s)
- Nitin Wankhade
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
| | - Anju Sharma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
| | - Mushtaq Ahmad Wani
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
| | - Aritra Banerjee
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
| |
Collapse
|
5
|
Romeo PH, Conquet L, Messiaen S, Pascal Q, Moreno SG, Bravard A, Bernardino-Sgherri J, Dereuddre-Bosquet N, Montagutelli X, Le Grand R, Petit V, Ferri F. Multiple Mechanisms of Action of Sulfodyne ®, a Natural Antioxidant, against Pathogenic Effects of SARS-CoV-2 Infection. Antioxidants (Basel) 2024; 13:1083. [PMID: 39334742 PMCID: PMC11429452 DOI: 10.3390/antiox13091083] [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: 07/10/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Few therapeutic options are available to treat COVID-19. The KEAP1/NRF2 pathway, the major redox-responsive pathway, has emerged as a potential therapeutic target for COVID-19 as it regulates redox homeostasis and inflammation that are altered during SARS-CoV-2 infection. Here, we characterized the effects of NRF2-agonist Sulfodyne®, a stabilized natural Sulforaphane, in cellular and animal models of SARS-CoV-2 infection. In pulmonary or colonic epithelial cell lines, Sulfodyne® elicited a more efficient inhibition of SARS-CoV-2 replication than NRF2-agonists DMF and CDDO. This antiviral activity was not dependent on NRF2 but was associated with the regulation of several metabolic pathways, including the inhibition of ER stress and mTOR signaling, which are activated during SARS-CoV-2 infection. Sulfodyne® also decreased SARS-CoV-2 mediated inflammatory responses by inhibiting the delayed induction of IFNB1 and type I IFN-stimulated genes in infected epithelial cell lines and by reducing the activation of human by-stander monocytes recruited after SARS-CoV-2 infection. In K18-hACE2 mice infected with SARS-CoV-2, Sulfodyne® treatment reduced both early lung viral load and disease severity by fine-tuning IFN-beta levels. Altogether, these results provide evidence for multiple mechanisms that underlie the antiviral and anti-inflammatory activities of Sulfodyne® and pinpoint Sulfodyne® as a potent therapeutic agent against pathogenic effects of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Paul-Henri Romeo
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Laurine Conquet
- Mouse Genetics Laboratory, Université Paris Cité, Institut Pasteur, 75724 Paris, France
| | - Sébastien Messiaen
- Laboratory on Development of the Gonads (LDG/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Development of the Gonads (LDG/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Quentin Pascal
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Stéphanie G Moreno
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Anne Bravard
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Jacqueline Bernardino-Sgherri
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Xavier Montagutelli
- Mouse Genetics Laboratory, Université Paris Cité, Institut Pasteur, 75724 Paris, France
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Vanessa Petit
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| | - Federica Ferri
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris Cité, Inserm, CEA, 92265 Fontenay-aux-Roses, France
- Laboratory on Repair and Transcription in Hematopoietic Stem Cells (LRTS/IRCM), Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
| |
Collapse
|
6
|
Lucaj T, Hay I, Samarbakhsh A, Bedi M, Iyer AK, Gavande NS. An overview of the development of pharmacotherapeutics targeting SARS-CoV-2. Drug Discov Today 2024; 29:104126. [PMID: 39097220 DOI: 10.1016/j.drudis.2024.104126] [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: 01/06/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Coronavirus disease 2019 (COVID-19) was declared a global pandemic in March 2020, which precipitated urgent public health responses. The causative agent, SARS-CoV-2, spreads primarily via respiratory droplets, necessitating precautions to mitigate transmission risks. Biopharmaceutical industries and academic institutions worldwide swiftly redirected their research endeavors towards developing therapeutic interventions, focusing on monoclonal antibodies, antiviral agents, and immunomodulatory therapies. The evolving body of evidence surrounding these treatments has prompted successive updates and revisions from the FDA, delineating the evolving landscape of COVID-19 therapeutics. This review comprehensively examines each treatment modality within the context of their developmental trajectories and regulatory approvals throughout the pandemic. Furthermore, it elucidates their mechanisms of action and presents clinical data underpinning their utility in combating the COVID-19 crisis.
Collapse
Affiliation(s)
- Tom Lucaj
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Ian Hay
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Mel Bedi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
7
|
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
|
8
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int J Mol Sci 2024; 25:7209. [PMID: 39000315 PMCID: PMC11241800 DOI: 10.3390/ijms25137209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Aprotinin is a broad-spectrum inhibitor of human proteases that has been approved for the treatment of bleeding in single coronary artery bypass surgery because of its potent antifibrinolytic actions. Following the outbreak of the COVID-19 pandemic, there was an urgent need to find new antiviral drugs. Aprotinin is a good candidate for therapeutic repositioning as a broad-spectrum antiviral drug and for treating the symptomatic processes that characterise viral respiratory diseases, including COVID-19. This is due to its strong pharmacological ability to inhibit a plethora of host proteases used by respiratory viruses in their infective mechanisms. The proteases allow the cleavage and conformational change of proteins that make up their viral capsid, and thus enable them to anchor themselves by recognition of their target in the epithelial cell. In addition, the activation of these proteases initiates the inflammatory process that triggers the infection. The attraction of the drug is not only its pharmacodynamic characteristics but also the possibility of administration by the inhalation route, avoiding unwanted systemic effects. This, together with the low cost of treatment (≈2 Euro/dose), makes it a good candidate to reach countries with lower economic means. In this article, we will discuss the pharmacodynamic, pharmacokinetic, and toxicological characteristics of aprotinin administered by the inhalation route; analyse the main advances in our knowledge of this medication; and the future directions that should be taken in research in order to reposition this medication in therapeutics.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
9
|
Schwartz J, Capistrano KJ, Gluck J, Hezarkhani A, Naqvi AR. SARS-CoV-2, periodontal pathogens, and host factors: The trinity of oral post-acute sequelae of COVID-19. Rev Med Virol 2024; 34:e2543. [PMID: 38782605 PMCID: PMC11260190 DOI: 10.1002/rmv.2543] [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: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
COVID-19 as a pan-epidemic is waning but there it is imperative to understand virus interaction with oral tissues and oral inflammatory diseases. We review periodontal disease (PD), a common inflammatory oral disease, as a driver of COVID-19 and oral post-acute-sequelae conditions (PASC). Oral PASC identifies with PD, loss of teeth, dysgeusia, xerostomia, sialolitis-sialolith, and mucositis. We contend that PD-associated oral microbial dysbiosis involving higher burden of periodontopathic bacteria provide an optimal microenvironment for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These pathogens interact with oral epithelial cells activate molecular or biochemical pathways that promote viral adherence, entry, and persistence in the oral cavity. A repertoire of diverse molecules identifies this relationship including lipids, carbohydrates and enzymes. The S protein of SARS-CoV-2 binds to the ACE2 receptor and is activated by protease activity of host furin or TRMPSS2 that cleave S protein subunits to promote viral entry. However, PD pathogens provide additional enzymatic assistance mimicking furin and augment SARS-CoV-2 adherence by inducing viral entry receptors ACE2/TRMPSS, which are poorly expressed on oral epithelial cells. We discuss the mechanisms involving periodontopathogens and host factors that facilitate SARS-CoV-2 infection and immune resistance resulting in incomplete clearance and risk for 'long-haul' oral health issues characterising PASC. Finally, we suggest potential diagnostic markers and treatment avenues to mitigate oral PASC.
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Oral Medicine and Diagnostic Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | | | - Joseph Gluck
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Armita Hezarkhani
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Afsar R. Naqvi
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| |
Collapse
|
10
|
Alemu J, Gumi B, Tsegaye A, Rahimeto Z, Fentahun D, Ibrahim F, Abubeker A, Gebremedhin A, Gelanew T, Howe R. Seroprevalence of SARS-CoV-2 and Hepatitis B Virus Coinfections among Ethiopians with Acute Leukemia. Cancers (Basel) 2024; 16:1606. [PMID: 38672687 PMCID: PMC11049053 DOI: 10.3390/cancers16081606] [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: 02/03/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
SARS-CoV-2 and blood-borne viral coinfections are well reported. Nevertheless, little is known regarding the seroprevalence of SARS-CoV-2 and coinfection with blood-borne viruses in hematologic malignancy patients in Ethiopia. We aimed to assess the seroprevalence of SARS-CoV-2 and associated infections with hepatitis B and other viruses among adolescent and adult acute leukemia patients at Tikur Anbessa Specialized Hospital, Addis Ababa, Ethiopia. A cross-sectional study was conducted from July 2020 to June 2021. Blood samples were tested for the presence of anti-SARS-CoV-2, HBV, HCV, and HIV with ELISA kits and occult hepatitis B infection with a real-time polymerase chain reaction assay. Out of a total 110 cases, the SARS-CoV-2 seroprevalence was 35.5%. The prevalence showed a significant increment from July 2020 to the end of June 2021 (p = 0.015). In 22.7% and 2.7% of leukemia cases, HBV and HIV, respectively, were detected. No HCV was identified. The rate of SARS-CoV-2 coinfection with HBV and HIV was 28% (11/39) and 2.6% (1/39), respectively; however, there was no statistically significant association between SARS-CoV-2 seropositivity with HBV and HIV (p > 0.05). There is a need for viral screening in leukemia cases to monitor infections and inform management.
Collapse
Affiliation(s)
- Jemal Alemu
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia;
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia;
| | - Balako Gumi
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia;
| | - Aster Tsegaye
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia;
| | - Ziyada Rahimeto
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
| | - Dessalegn Fentahun
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
| | - Fozia Ibrahim
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
| | - Abdulaziz Abubeker
- Department of Internal Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (A.A.); (A.G.)
| | - Amha Gebremedhin
- Department of Internal Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (A.A.); (A.G.)
| | - Tesfaye Gelanew
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
| | - Rawleigh Howe
- Armauer Hansen Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (Z.R.); (D.F.); (F.I.); (T.G.); (R.H.)
| |
Collapse
|
11
|
Xu F, Qin Y, Guan C. Discovery of components in honeysuckle for treating COVID-19 and diabetes based on molecular docking, network analysis and experimental validation. Nat Prod Res 2024:1-5. [PMID: 38591097 DOI: 10.1080/14786419.2024.2340040] [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: 11/06/2023] [Accepted: 03/27/2024] [Indexed: 04/10/2024]
Abstract
Molecular docking screening identified ochnaflavone, madreselvin B and hydnocarpin as key components for treating COVID-19 with diabetes in honeysuckle using 3 C-like protease (Mpro), angiotensin-converting enzyme 2 (ACE2), and dipeptidyl peptidase 4 (DPP4) as molecular docking targets, ACE2, DPP4, IL2, NFKB1, PLG, TBK1, TLR4 and TNF were the core targets, and multiple antiviral and anti-inflammatory signalling pathways were involved. Further, the levels of IL-1β and DPP4 in cell supernatant that had been activated by LPS was decreased by hypnocarpin, and ACE2 protein and DPP4 mRNA in cells were down-regulated. Overall, we have identified three components from honeysuckle that have potency to treat COVID-19 combined with diabetes. SARS-CoV-2 transcription may be inhibited by these components in honeysuckle, reducing virus invasion, inhibiting inflammatory factors, and improving immune response. Our findings could provide a basis for the clinical application and further development of honeysuckle.
Collapse
Affiliation(s)
- Feng Xu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ya Qin
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Changxiu Guan
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
12
|
Steenblock C, Richter S, Lindemann D, Ehrlich H, Bornstein SR, Bechmann N. Marine Sponge-Derived Secondary Metabolites Modulate SARS-CoV-2 Entry Mechanisms. Horm Metab Res 2024; 56:308-317. [PMID: 37793428 DOI: 10.1055/a-2173-0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
The emergence of SARS-CoV 2 caused the COVID-19 pandemic, resulting in numerous global infections and deaths. In particular, people with metabolic diseases display an increased risk of severe COVID 19 and a fatal outcome. Treatment options for severe cases are limited, and the appearance of new virus variants complicates the development of novel therapies. To better manage viral infections like COVID 19, new therapeutic approaches are needed. Marine sponges offer a natural and renewable source of unique bioactive agents. These sponges produce secondary metabolites with various effects, including anti-viral, anti-inflammatory, and anti-tumorigenic properties. In the current study, we investigated the effect of five different marine sponge-derived secondary metabolites (four bromotyrosines and one sesquiterpenoid hydroquinone). Two of these, Avarol and Acetyl-dibromoverongiaquinol reduced the expression of ACE2, the main receptor for SARS-CoV 2, and the alternative receptor NRP1. Moreover, these substances derived from sponges demonstrated the ability to diminish the virus titer in SARS-CoV 2-infected cells, especially concerning the Omicron lineage. However, the reduction was not substantial enough to expect a significant impact on infected humans. Consequently, the investigated sponge-derived secondary metabolites are not likely to be effective to treat COVID 19 as a stand-alone therapy.
Collapse
Affiliation(s)
- Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefanie Richter
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Dirk Lindemann
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hermann Ehrlich
- Center for Advanced Technologies, Adam Mickiewicz University, Poznan, Poland
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
13
|
Mora-Rodríguez JM, Sánchez BG, Bort A, Díaz-Yuste A, Ballester-González R, Arrieta F, Sebastián-Martín A, Díaz-Laviada I. Diabetic individuals with COVID-19 exhibit reduced efficacy of gliptins in inhibiting dipeptidyl peptidase 4 (DPP4). A suggested explanation for increased COVID-19 susceptibility in patients with type 2 diabetes mellitus (T2DM). Life Sci 2024; 336:122292. [PMID: 38030058 DOI: 10.1016/j.lfs.2023.122292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
AIMS Dipeptidyl peptidase 4 (DPP4) has been proposed as a coreceptor for SARS-CoV-2 cellular entry. Considering that type 2 diabetes mellitus (T2DM) has been identified as the most important risk factor for SARS-CoV-2, and that gliptins (DPP4 inhibitors) are a prescribed diabetic treatment, this study aims to unravel the impact of DPP4 in the intersection of T2DM/COVID-19. MATERIALS AND METHODS We analyzed 189 serum human samples, divided into six clinical groups (controls, T2DM, T2DM + gliptins, COVID-19, COVID-19 + T2DM, and COVID-19 + T2DM + gliptins), measuring DPP4 protein concentration and activity by Western blot, ELISA, and commercial activity kits. The obtained results were verified in Huh-7 cellular models. KEY FINDINGS Both DPP4 concentration and activity were decreased in COVID-19 patients, and as in T2DM patients, compared to controls. Despite these lower levels, the ratio of DPP4 activity/concentration in COVID-19 sera was the highest (0.782 ± 0.289 μU/ng vs. 0.547 ± 0.050 μU/ng in controls, p < 0.0001), suggesting a compensating mechanism in these patients. Supernatants of Huh-7 cells incubated with COVID-19 serum showed a consistent and significantly lower DPP4 concentration and activity. Furthermore, COVID-19 + T2DM + gliptins patients showed a higher serum DPP4 concentration and activity than T2DM + gliptin subjects (p < 0.05), indicating that sera from COVID-19 convalescents interfere with gliptins. SIGNIFICANCE Either SARS-CoV-2 or some metabolites present in the sera of COVID-19-convalescent patients interact with soluble DPP4 or even gliptins themselves since the inhibitory effect of gliptins on DPP4 activity is being prevented. The interactions between DPP4, gliptins, and SARS-CoV-2 should be further elucidated to reveal the mechanism of action for these interesting observations.
Collapse
Affiliation(s)
- José María Mora-Rodríguez
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain.
| | - Belén G Sánchez
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain.
| | - Alicia Bort
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain.
| | - Alba Díaz-Yuste
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain.
| | - Rubén Ballester-González
- Immunology Service, Ramón y Cajal Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain.
| | - Francisco Arrieta
- Endocrinology and Nutrition Service, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain.
| | - Alba Sebastián-Martín
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain.
| | - Inés Díaz-Laviada
- Department of Systems Biology, School of Medicine and Health Sciences, Alcalá University, Alcalá de Henares, Spain; Health Research Institute of Castilla-La Mancha (IDISCAM), Spain; Chemical Research Institute "Andrés M. del Río" (IQAR), Alcalá de Henares, Spain.
| |
Collapse
|
14
|
Hubáček JA, Philipp T, Adámková V, Májek O, Dlouhá D, Dušek L. Possible effect of OAS1 and TMPRSS6 but not DPP4 and ZNF335 polymorphisms on COVID-19 severity in the Czech population. Cent Eur J Public Health 2023; 31:235-239. [PMID: 38309700 DOI: 10.21101/cejph.a7906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 02/05/2024]
Abstract
OBJECTIVES The acute respiratory syndrome, known as COVID-19, is characterised by high morbidity and increased mortality. Genetic factors may partially explain the differences in susceptibility to and severity of COVID-19. METHODS We have analysed common functional polymorphisms within the OAS1 (rs4767027), TMPRSS6 (rs855791), DPP4 (rs3788979), and ZNF335 (rs3848719) genes in SARS-CoV-2 positive subjects (n = 521, different disease severity) and in population controls (n = 2,559 subjects, COVID-19 status unknown). RESULTS Neither DPP4 nor ZNF335 were associated with disease susceptibility or severity in the Czech population in any of the models used for calculation. T allele carriers of the OAS1 polymorphism seem to be protective against symptomatic COVID-19 (p = 0.002 calculated for trend; asymptomatic, symptomatic, hospitalised). Similarly, within the TMPRSS6, minor TT homozygotes associated with lower plasma Fe concentrations were underrepresented in the overall patient group (p = 0.044; OR = 0.77, 95% CI: 0.59-0.99), and the difference was mainly driven by the severe COVID-19 subjects. In general, risky homozygotes of these two polymorphisms were less frequent than expected in the group of hospitalised COVID-19 survivors. CONCLUSIONS Common variants within OAS1 (rs4767027) and TMPRSS6 (rs855791) play some role in COVID-19 pathology in the Czech Caucasian population. Whether the depletion of minor allele carriers of these two variants is associated with increased COVID-19 mortality, needs to be analysed in an external confirmatory study.
Collapse
Affiliation(s)
- Jaroslav A Hubáček
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Endocrinology and Metabolism, Third Department of Internal Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tom Philipp
- Clinic of Rheumatology and Physiotherapy, Third Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czech Republic
| | - Věra Adámková
- Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondřej Májek
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dana Dlouhá
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ladislav Dušek
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
15
|
Rohani, Febrina E, Wahyuni IS, Levita J. Pharmacological and Clinical Studies of Medicinal Plants That Inhibit Dipeptidyl Peptidase-IV. Drug Des Devel Ther 2023; 17:3473-3491. [PMID: 38024536 PMCID: PMC10680473 DOI: 10.2147/dddt.s426870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Dipeptidyl peptidase IV (DPP-IV) is an enzyme responsible for the degradation of the incretin hormone glucagon-like peptide-1 (GLP-1). DPP-IV plays a significant role in regulating blood glucose levels by modulating the activity of GLP-1. In the context of diabetes, DPP-IV inhibitors effectively block the activity of DPP-IV, hence mitigating the degradation of GLP-1. This, in turn, leads to an extension of GLP-1's duration of action, prolongs gastric emptying, enhances insulin sensitivity, and ultimately results in the reduction of blood glucose levels. Nonetheless, reported adverse events of DPP-IV inhibitors on T2DM patients make it essential to understand the activity and mechanism of these drugs, particularly viewed from the perspective of finding the effective and safe add-on medicinal plants, to be implemented in clinical practice. This review is intended to bring forth a thorough overview of plants that work by reducing DPP-IV activity, from computational technique, enzymatic study, animal experiments, and studies in humans. The articles were searched on PubMed using "Plants", "DPP-IV", "DPP-IV inhibitor", "GLP-1", "Type 2 diabetes", "diabetes", "in silico", "in vitro", "in vivo", "studies in human", "clinical study" as the query words, and filtered for ten years of publication period. Eighteen plants showed inhibition against DPP-IV as proven by in silico, in vitro, and in vivo studies; however, only ten plants were reported for efficacy in clinical studies. Several plant-based DPP-IV inhibitors, eg, Allium sativum, Morus Alba, Curcuma longa, Pterocarpus marsupium, and Taraxacum officinale, have established their functional role in inhibiting DPP-IV and have proven their effectiveness through studies in humans earning them a prominent place in therapeutic discovery.
Collapse
Affiliation(s)
- Rohani
- Master Program in Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| | - Ellin Febrina
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| | - Indah Suasani Wahyuni
- Department of Oral Medicine, Faculty of Dentistry, Padjadjaran University, Sumedang, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| |
Collapse
|
16
|
Ghorbanali Z, Zare-Mirakabad F, Salehi N, Akbari M, Masoudi-Nejad A. DrugRep-HeSiaGraph: when heterogenous siamese neural network meets knowledge graphs for drug repurposing. BMC Bioinformatics 2023; 24:374. [PMID: 37789314 PMCID: PMC10548718 DOI: 10.1186/s12859-023-05479-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Drug repurposing is an approach that holds promise for identifying new therapeutic uses for existing drugs. Recently, knowledge graphs have emerged as significant tools for addressing the challenges of drug repurposing. However, there are still major issues with constructing and embedding knowledge graphs. RESULTS This study proposes a two-step method called DrugRep-HeSiaGraph to address these challenges. The method integrates the drug-disease knowledge graph with the application of a heterogeneous siamese neural network. In the first step, a drug-disease knowledge graph named DDKG-V1 is constructed by defining new relationship types, and then numerical vector representations for the nodes are created using the distributional learning method. In the second step, a heterogeneous siamese neural network called HeSiaNet is applied to enrich the embedding of drugs and diseases by bringing them closer in a new unified latent space. Then, it predicts potential drug candidates for diseases. DrugRep-HeSiaGraph achieves impressive performance metrics, including an AUC-ROC of 91.16%, an AUC-PR of 90.32%, an accuracy of 84.63%, a BS of 0.119, and an MCC of 69.31%. CONCLUSION We demonstrate the effectiveness of the proposed method in identifying potential drugs for COVID-19 as a case study. In addition, this study shows the role of dipeptidyl peptidase 4 (DPP-4) as a potential receptor for SARS-CoV-2 and the effectiveness of DPP-4 inhibitors in facing COVID-19. This highlights the practical application of the model in addressing real-world challenges in the field of drug repurposing. The code and data for DrugRep-HeSiaGraph are publicly available at https://github.com/CBRC-lab/DrugRep-HeSiaGraph .
Collapse
Affiliation(s)
- Zahra Ghorbanali
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Fatemeh Zare-Mirakabad
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran.
| | - Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Mohammad Akbari
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
17
|
Yazdanparast S, Bakhtiyaridovvombaygi M, Mikanik F, Ahmadi R, Ghorbani M, Mansoorian MR, Mansoorian M, Chegni H, Moshari J, Gharehbaghian A. Spotlight on contributory role of host immunogenetic profiling in SARS-CoV-2 infection: Susceptibility, severity, mortality, and vaccine effectiveness. Life Sci 2023:121907. [PMID: 37394094 DOI: 10.1016/j.lfs.2023.121907] [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: 03/04/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND The SARS-CoV-2 virus has spread continuously worldwide, characterized by various clinical symptoms. The immune system responds to SARS-CoV-2 infection by producing Abs and secreting cytokines. Recently, numerous studies have highlighted that immunogenetic factors perform a putative role in COVID-19 pathogenesis and implicate vaccination effectiveness. AIM This review summarizes the relevant articles and evaluates the significance of mutation and polymorphism in immune-related genes regarding susceptibility, severity, mortality, and vaccination effectiveness of COVID-19. Furthermore, the correlation between host immunogenetic and SARS-CoV-2 reinfection is discussed. METHOD A comprehensive search was conducted to identify relevant articles using five databases until January 2023, which resulted in 105 total articles. KEY FINDINGS Taken to gather this review summarized that: (a) there is a plausible correlation between immune-related genes and COVID-19 outcomes, (b) the HLAs, cytokines, chemokines, and other immune-related genes expression profiles can be a prognostic factor in COVID-19-infected patients, and (c) polymorphisms in immune-related genes have been associated with the effectiveness of vaccination. SIGNIFICANCE Regarding the importance of mutation and polymorphisms in immune-related genes in COVID-19 outcomes, modulating candidate genes is expected to help clinical decisions, patient outcomes management, and innovative therapeutic approach development. In addition, the manipulation of host immunogenetics is hypothesized to induce more robust cellular and humoral immune responses, effectively increase the efficacy of vaccines, and subsequently reduce the incidence rates of reinfection-associated COVID-19.
Collapse
Affiliation(s)
- Somayeh Yazdanparast
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Ahmadi
- Department of Infectious Diseases, School of Medicine, Infectious Diseases Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Mohammad Ghorbani
- Laboratory Hematology and Transfusion Medicine, Department of Pathology, Faculty Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| | | | - Mozhgan Mansoorian
- Nursing Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Hamid Chegni
- Department of Immunology, School of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalil Moshari
- School of Medicine, Gonabad University of Medical Science, Gonabad, Iran
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Mani S, Kaur A, Jakhar K, Kumari G, Sonar S, Kumar A, Das S, Kumar S, Kumar V, Kundu R, Pandey AK, Singh UP, Majumdar T. Targeting DPP4-RBD interactions by sitagliptin and linagliptin delivers a potential host-directed therapy against pan-SARS-CoV-2 infections. Int J Biol Macromol 2023; 245:125444. [PMID: 37385308 PMCID: PMC10293653 DOI: 10.1016/j.ijbiomac.2023.125444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Highly mutated SARS-CoV-2 is known aetiological factor for COVID-19. Here, we have demonstrated that the receptor binding domain (RBD) of the spike protein can interact with human dipeptidyl peptidase 4 (DPP4) to facilitate virus entry, in addition to the usual route of ACE2-RBD binding. Significant number of residues of RBD makes hydrogen bonds and hydrophobic interactions with α/β-hydrolase domain of DPP4. With this observation, we created a strategy to combat COVID-19 by circumventing the catalytic activity of DPP4 using its inhibitors. Sitagliptin, linagliptin or in combination disavowed RBD to establish a heterodimer complex with both DPP4 and ACE2 which is requisite strategy for virus entry into the cells. Both gliptins not only impede DPP4 activity, but also prevent ACE2-RBD interaction, crucial for virus growth. Sitagliptin, and linagliptin alone or in combination have avidity to impede the growth of pan-SARS-CoV-2 variants including original SARS-CoV-2, alpha, beta, delta, and kappa in a dose dependent manner. However, these drugs were unable to alter enzymatic activity of PLpro and Mpro. We conclude that viruses hijack DPP4 for cell invasion via RBD binding. Impeding RBD interaction with both DPP4 and ACE2 selectively by sitagliptin and linagliptin is an potential strategy for efficiently preventing viral replication.
Collapse
Affiliation(s)
- Shailendra Mani
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Kamini Jakhar
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Sudipta Sonar
- Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Kumar
- National Institute of Immunology, New Delhi, India
| | - Sudesna Das
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | | - Vijay Kumar
- National Institute of Immunology, New Delhi, India
| | - Rakesh Kundu
- Department of Zoology, Visva-Bharati University, Santiniketan, West Bengal, India
| | - Anil Kumar Pandey
- Department of Physiology, ESIC Medical College & Hospital, Faridabad, India
| | | | | |
Collapse
|
19
|
Barkhordarian M, Behbood A, Ranjbar M, Rahimian Z, Prasad A. Overview of the cardio-metabolic impact of the COVID-19 pandemic. Endocrine 2023; 80:477-490. [PMID: 37103684 PMCID: PMC10133915 DOI: 10.1007/s12020-023-03337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 04/28/2023]
Abstract
Evidence has shown that cardiometabolic disorders (CMDs) are amongst the top contributors to COVID-19 infection morbidity and mortality. The reciprocal impact of COVID-19 infection and the most common CMDs, the risk factors for poor composite outcome among patients with one or several underlying diseases, the effect of common medical management on CMDs and their safety in the context of acute COVID-19 infection are reviewed. Later on, the changes brought by the COVID-19 pandemic quarantine on the general population's lifestyle (diet, exercise patterns) and metabolic health, acute cardiac complications of different COVID-19 vaccines and the effect of CMDs on the vaccine efficacy are discussed. Our review identified that the incidence of COVID-19 infection is higher among patients with underlying CMDs such as hypertension, diabetes, obesity and cardiovascular disease. Also, CMDs increase the risk of COVID-19 infection progression to severe disease phenotypes (e.g. hospital and/or ICU admission, use of mechanical ventilation). Lifestyle modification during COVID-19 era had a great impact on inducing and worsening of CMDs. Finally, the lower efficacy of COVID-19 vaccines was found in patients with metabolic disease.
Collapse
Affiliation(s)
- Maryam Barkhordarian
- Department of Medicine, Division of Cardiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Arezoo Behbood
- MPH department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Maryam Ranjbar
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Zahra Rahimian
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Anand Prasad
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
20
|
Zahedi M, Kordrostami S, Kalantarhormozi M, Bagheri M. A Review of Hyperglycemia in COVID-19. Cureus 2023; 15:e37487. [PMID: 37187644 PMCID: PMC10181889 DOI: 10.7759/cureus.37487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetes mellitus (DM) is one of the most common chronic metabolic disorders worldwide, which increases the risk of common and opportunistic infections. Following the coronavirus disease 2019 (COVID-19) pandemic, a higher incidence rate, more severe forms of the disease, and exacerbation of hyperglycemia and its complications have been observed in patients with DM. Moreover, stress-induced hyperglycemia has been observed in many hospitalized nondiabetic patients after contracting COVID-19. Hyperglycemia worsens prognosis in both diabetic and nondiabetic patients. In this study, the mechanism of new-onset or exacerbation of hyperglycemia, the effect of the treatments used for COVID-19 on hyperglycemia, the importance and appropriate method of blood glucose (blood sugar (BS)) control during the disease, and the possible fate of new-onset hyperglycemia after recovery from COVID-19 to some extent is expressed.
Collapse
Affiliation(s)
- Maryam Zahedi
- Department of Internal Medicine, Endocrinology, and Metabolism, Clinical Research Development Unit (CRDU) 5 Azar Hospital, Golestan University of Medical Sciences, Gorgan, IRN
| | - Saba Kordrostami
- Department of Endocrinology and Diabetes, Clinical Research Development Unit (CRDU) 5 Azar Hospital, Golestan University of Medical Sciences, Gorgan, IRN
| | | | - Marziyeh Bagheri
- Department of Internal Medicine, Bushehr University of Medical Sciences, Bushehr, IRN
| |
Collapse
|
21
|
Nag S, Mandal S, Mukherjee O, Mukherjee S, Kundu R. DPP-4 Inhibitors as a savior for COVID-19 patients with diabetes. Future Virol 2023:10.2217/fvl-2022-0112. [PMID: 37064327 PMCID: PMC10096336 DOI: 10.2217/fvl-2022-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 03/01/2023] [Indexed: 04/18/2023]
Abstract
Diabetic patients are at particular risk of severe COVID-19. Human dipeptidyl peptidase-4 (DPP-4) is a membrane-bound aminopeptidase that regulates insulin release by inactivating incretin. DPP-4 inhibitors (DPP-4is) are therefore used as oral anti-diabetic drugs to restore normal insulin levels. These molecules also have anti-inflammatory and anti-hypertension effects. Recent studies on the interactions of SARS-CoV-2 spike glycoprotein and DPP-4 predict a possible entry route for SARS-CoV-2. Therefore, DPP-4is could be effective at reducing the virus-induced 'cytokine storm', thereby ceasing inflammatory injury to vital organs. Moreover, DPP-4is may interfere with viral entry into host cells. Herein, we have reviewed the efficacy of DPP-4is as potential repurposed drugs to reduce the severity of SARS-CoV-2 infection in patients with diabetes.
Collapse
Affiliation(s)
- Snehasish Nag
- Department of Zoology, Cell Signaling Laboratory, Visva-Bharati University, Santiniketan, West Bengal, 731 235, India
| | - Samanwita Mandal
- Department of Zoology, Cell Signaling Laboratory, Visva-Bharati University, Santiniketan, West Bengal, 731 235, India
| | - Oindrila Mukherjee
- Department of Zoology, Cell Signaling Laboratory, Visva-Bharati University, Santiniketan, West Bengal, 731 235, India
| | - Suprabhat Mukherjee
- Department of Animal Science, Integrative Biochemistry & Immunology Laboratory, Kazi Nazrul University, Asansol, West Bengal, 713 340, India
- Author for correspondence:
| | - Rakesh Kundu
- Department of Zoology, Cell Signaling Laboratory, Visva-Bharati University, Santiniketan, West Bengal, 731 235, India
- Author for correspondence:
| |
Collapse
|
22
|
Du J, Fu J, Zhang W, Zhang L, Chen H, Cheng J, He T, Fu J. Effect of DPP4/CD26 expression on SARS‑CoV‑2 susceptibility, immune response, adenosine (derivatives m 62A and CD) regulations on patients with cancer and healthy individuals. Int J Oncol 2023; 62:41. [PMID: 36799191 PMCID: PMC9946808 DOI: 10.3892/ijo.2023.5489] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
The worldwide COVID‑19 pandemic was brought on by a new coronavirus (SARS Cov‑2). A marker/receptor called Dipeptidyl peptidase 4/CD26(DPP4/CD26) may be crucial in determining susceptibility to tumors and coronaviruses. However, the regulation of DPP4 in COVID‑invaded cancer patients and its role on small molecule compounds remain unclear. The present study used the Human Protein Atlas, Monaco, and Schmiedel databases to analyze the expression of DPP4 in human tissues and immune cells. The association between DPP4 expression and survival in various tumor tissues was compared using GEPIA 2. The DNMIVD database was used to analyze the correlation between DPP4 expression and promoter methylation in various tumors. On the cBioPortal network, the frequency of DPP4 DNA mutations in various cancers was analyzed. The correlation between DPP4 expression and immunomodulators was analyzed by TISIDB database. The inhibitory effects of cordycepin (CD), N6, N6‑dimethyladenosine (m62A) and adenosine (AD) on DPP4 in cancer cells were evaluated. DPP4 was mainly expressed in endocrine tissue, followed by gastrointestinal tract, female tissue (mainly in placenta), male tissue (mainly in prostate and seminal vesicle), proximal digestive tract, kidney, bladder, liver, gallbladder and respiratory system. In immune cells, DPP4 mRNA was mainly expressed in T cells, and its expression was upregulated in esophageal carcinoma, kidney renal papillary cell carcinoma (KIRP), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma, pancreatic adenocarcinoma, prostate adenocarcinoma, stomach adenocarcinoma, thyroid carcinoma and thymoma. However, it was downregulated in breast invasive carcinoma, kidney chromophobe, lung squamous cell carcinoma and skin cutaneous melanoma. Thus, DPP4 is involved in viral invasion in most types of cancer. The expression of DPP4 could be inhibited by CD, m62A and AD in different tumor cells. Moreover, CD significantly inhibited the formation of GFP‑positive syncytial cells. In vivo experiments with AD injection further showed that AD significantly inhibited lymphocyte activating factor 3 expression. These drugs may have potential to treat COVID‑19 by targeting DPP4. Thus, DPP4 may be medically significant for SARS‑CoV‑2‑infected cancer patients, providing prospective novel targets and concepts for the creation of drugs against COVID‑19.
Collapse
Affiliation(s)
- Jiaman Du
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenqian Zhang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lianmei Zhang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China,Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Hanchun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao He
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China,Institute for Cancer Medicine and Basic Medical School, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China,Correspondence to: Professor Junjiang Fu or Professor Tao He, Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, Sichuan 646000, P.R. China, E-mail: , E-mail:
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China,Correspondence to: Professor Junjiang Fu or Professor Tao He, Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, Sichuan 646000, P.R. China, E-mail: , E-mail:
| |
Collapse
|
23
|
Garmendia JV, García AH, De Sanctis CV, Hajdúch M, De Sanctis JB. Autoimmunity and Immunodeficiency in Severe SARS-CoV-2 Infection and Prolonged COVID-19. Curr Issues Mol Biol 2022; 45:33-50. [PMID: 36661489 PMCID: PMC9857622 DOI: 10.3390/cimb45010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2 causes the complex and heterogeneous illness known as COVID-19. The disease primarily affects the respiratory system but can quickly become systemic, harming multiple organs and leading to long-lasting sequelae in some patients. Most infected individuals are asymptomatic or present mild symptoms. Antibodies, complement, and immune cells can efficiently eliminate the virus. However, 20% of individuals develop severe respiratory illness and multiple organ failure. Virus replication has been described in several organs in patients who died from COVID-19, suggesting a compromised immune response. Immunodeficiency and autoimmunity are responsible for this impairment and facilitate viral escape. Mutations in IFN signal transduction and T cell activation are responsible for the inadequate response in young individuals. Autoantibodies are accountable for secondary immunodeficiency in patients with severe infection or prolonged COVID-19. Antibodies against cytokines (interferons α, γ and ω, IL1β, IL6, IL10, IL-17, IL21), chemokines, complement, nuclear proteins and DNA, anticardiolipin, and several extracellular proteins have been reported. The type and titer of autoantibodies depend on age and gender. Organ-specific autoantibodies have been described in prolonged COVID-19. Their role in the disease is under study. Autoimmunity and immunodeficiency should be screened as risk factors for severe or prolonged COVID-19.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Alexis Hipólito García
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
- Czech Institute of Advanced Technology in Research [Catrin], Palacky University, 779 00 Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
- Czech Institute of Advanced Technology in Research [Catrin], Palacky University, 779 00 Olomouc, Czech Republic
- Correspondence:
| |
Collapse
|