1
|
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
|
2
|
Marrone L, Romano S, Albanese M, Giordano S, Morello A, Cimmino M, Di Giacomo V, Malasomma C, Romano MF, Corcione N. Tirofiban prevents the effects of SARS-CoV-2 spike protein on macrophage activation and endothelial cell death. Heliyon 2024; 10:e35341. [PMID: 39170431 PMCID: PMC11336627 DOI: 10.1016/j.heliyon.2024.e35341] [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: 08/22/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
SARS-CoV-2 viral-derived particles have been proposed to have a causal role in tissue inflammation. Macrophage is the culprit cell in the pathogenesis of destructive inflammatory response to the SARS-CoV-2 virus. We investigated whether the spike protein might play a role in perturbing the physiological process of resolution of inflammation. Using an in vitro model of M2 polarized macrophages, we found that recombinant spike protein produced typical M1 morphological features in these alternative differentiated cells. In the presence of spike, M2-macrophages lose their elongated morphology, become rounded and acquire a strong capability to stimulate lymphocyte activation and proliferation. Moreover, in M2 macrophages, spike activated the signal transducer and activator-1 (STAT1) the pivotal mediator of pro-inflammatory macrophages. We observed STAT1 activation also in endothelial cells cultured with recombinant spike, accompanied by Bax upregulation and cell death. Blockade of beta3 integrin with the RGD mimetic tirofiban reverted the spike-induced costimulatory effects on M2 macrophages. Also, tirofiban counteracted STAT1 and Bax activation in endothelial cells cultured with spike and reduced endothelial cell death. In conclusion, we found that some proinflammatory effects of the spike protein can involve the integrin pathway and provide elements supporting use of RGD mimetics against SARS-Cov-2.
Collapse
Affiliation(s)
- Laura Marrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Michele Albanese
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Salvatore Giordano
- Division of Cardiology, Department of Medical and Surgical Sciences, “Magna Graecia” University, Catanzaro, Italy
| | - Alberto Morello
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Michele Cimmino
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Valeria Di Giacomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Chiara Malasomma
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Nicola Corcione
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| |
Collapse
|
3
|
Datta G, Rezagholizadeh N, Hasler WA, Khan N, Chen X. SLC38A9 regulates SARS-CoV-2 viral entry. iScience 2024; 27:110387. [PMID: 39071889 PMCID: PMC11277692 DOI: 10.1016/j.isci.2024.110387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/13/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
SARS-CoV-2 viral entry into host cells depends on the cleavage of spike (S) protein into S1 and S2 proteins. Such proteolytic cleavage by furin results in the exposure of a multibasic motif on S1, which is critical for SARS-CoV-2 viral infection and transmission; however, how such a multibasic motif contributes to the infection of SARS-CoV-2 remains elusive. Here, we demonstrate that the multibasic motif on S1 is critical for its interaction with SLC38A9, an endolysosome-resident arginine sensor. SLC38A9 knockdown prevents S1-induced endolysosome de-acidification and blocks the S protein-mediated entry of pseudo-SARS-CoV-2 in Calu-3, U87MG, Caco-2, and A549 cells. Our findings provide a novel mechanism in regulating SARS-CoV-2 viral entry; S1 present in endolysosome lumen could interact with SLC38A9, which mediates S1-induced endolysosome de-acidification and dysfunction, facilitating the escape of SARS-CoV-2 from endolysosomes and enhancing viral entry.
Collapse
Affiliation(s)
- Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Neda Rezagholizadeh
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Wendie A. Hasler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| |
Collapse
|
4
|
Liu YS, Chen WL, Zeng YW, Li ZH, Zheng HL, Pan N, Zhao LY, Wang S, Chen SH, Jiang MH, Jin CC, Mi YC, Cai ZH, Fang XZ, Liu YJ, Liu L, Wang GL. Isaridin E Protects against Sepsis by Inhibiting Von Willebrand Factor-Induced Endothelial Hyperpermeability and Platelet-Endothelium Interaction. Mar Drugs 2024; 22:283. [PMID: 38921594 PMCID: PMC11204489 DOI: 10.3390/md22060283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Endothelial hyperpermeability is pivotal in sepsis-associated multi-organ dysfunction. Increased von Willebrand factor (vWF) plasma levels, stemming from activated platelets and endothelium injury during sepsis, can bind to integrin αvβ3, exacerbating endothelial permeability. Hence, targeting this pathway presents a potential therapeutic avenue for sepsis. Recently, we identified isaridin E (ISE), a marine-derived fungal cyclohexadepsipeptide, as a promising antiplatelet and antithrombotic agent with a low bleeding risk. ISE's influence on septic mortality and sepsis-induced lung injury in a mouse model of sepsis, induced by caecal ligation and puncture, is investigated in this study. ISE dose-dependently improved survival rates, mitigating lung injury, thrombocytopenia, pulmonary endothelial permeability, and vascular inflammation in the mouse model. ISE markedly curtailed vWF release from activated platelets in septic mice by suppressing vesicle-associated membrane protein 8 and soluble N-ethylmaleide-sensitive factor attachment protein 23 overexpression. Moreover, ISE inhibited healthy human platelet adhesion to cultured lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs), thereby significantly decreasing vWF secretion and endothelial hyperpermeability. Using cilengitide, a selective integrin αvβ3 inhibitor, it was found that ISE can improve endothelial hyperpermeability by inhibiting vWF binding to αvβ3. Activation of the integrin αvβ3-FAK/Src pathway likely underlies vWF-induced endothelial dysfunction in sepsis. In conclusion, ISE protects against sepsis by inhibiting endothelial hyperpermeability and platelet-endothelium interactions.
Collapse
Affiliation(s)
- Yao-Sheng Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Wen-Liang Chen
- Scientific Research Center, the Medical Interdisciplinary Science Research Center of Western Guangdong, College of Women and Children, the Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China;
| | - Yu-Wei Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Zhi-Hong Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Hao-Lin Zheng
- Division of Biosciences, University College London, London WC1E 6BT, UK;
| | - Ni Pan
- Department of Pharmacy, The Second Clinical College, Guangzhou Medical University, Guangzhou 510261, China;
| | - Li-Yan Zhao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Shu Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Sen-Hua Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510006, China; (S.-H.C.); (M.-H.J.)
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Ming-Hua Jiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510006, China; (S.-H.C.); (M.-H.J.)
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Chen-Chen Jin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Yu-Chen Mi
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Zhao-Hui Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Xin-Zhe Fang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| | - Yong-Jun Liu
- Guangdong Provincial Clinical Research Center of Critical Care Medicine, Guangzhou 510080, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510006, China; (S.-H.C.); (M.-H.J.)
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (Y.-S.L.); (Y.-W.Z.); (Z.-H.L.); (L.-Y.Z.); (S.W.); (C.-C.J.); (Y.-C.M.); (Z.-H.C.); (X.-Z.F.)
| |
Collapse
|
5
|
Lu RXZ, Zhao Y, Radisic M. The emerging role of heart-on-a-chip systems in delineating mechanisms of SARS-CoV-2-induced cardiac dysfunction. Bioeng Transl Med 2024; 9:e10581. [PMID: 38818123 PMCID: PMC11135153 DOI: 10.1002/btm2.10581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 06/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a major global health concern since its emergence in 2019, with over 680 million confirmed cases as of April 2023. While COVID-19 has been strongly associated with the development of cardiovascular complications, the specific mechanisms by which viral infection induces myocardial dysfunction remain largely controversial as studies have shown that the severe acute respiratory syndrome coronavirus-2 can lead to heart failure both directly, by causing damage to the heart cells, and indirectly, by triggering an inflammatory response throughout the body. In this review, we summarize the current understanding of potential mechanisms that drive heart failure based on in vitro studies. We also discuss the significance of three-dimensional heart-on-a-chip technology in the context of the current and future pandemics.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Yimu Zhao
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
| | - Milica Radisic
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of Chemical Engineering and Applied ChemistryUniversity of TorontoTorontoOntarioCanada
- Terence Donnelly Centre for Cellular & Biomolecular ResearchUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
6
|
Zhang XZ, Wang J, Tian WJ, You JL, Chi XJ, Wang XJ. Phospho-eIF4E stimulation regulates coronavirus entry by selective expression of cell membrane-residential factors. J Virol 2024; 98:e0194823. [PMID: 38299843 PMCID: PMC10878034 DOI: 10.1128/jvi.01948-23] [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: 12/14/2023] [Accepted: 12/31/2023] [Indexed: 02/02/2024] Open
Abstract
The eukaryotic translation initiation factor eIF4E can regulate cellular translation via phosphorylation on serine 209. In a recent study, by two rounds of TMT relative quantitative proteomics, we found that phosphorylated eIF4E (p-eIF4E) favors the translation of selected mRNAs, and the encoded proteins are mainly involved in ECM-receptor, focal adhesion, and PI3K-Akt signaling. The current paper is focused on the relationship between p-eIF4E and the downstream host cell proteins, and their presumed effect on efficient entry of PEDV. We found that the depletion of membrane-residential factor TSPAN3, CD63, and ITGB2 significantly inhibited viral invasion of PEDV, and reduced the entry of pseudotyped particles PEDV-pp, SARS-CoV-pp, and SARS-CoV-2-pp. The specific antibodies of TSPAN3, CD63, and ITGB2 blocked the adsorption of PEDV into host cells. Moreover, we detected that eIF4E phosphorylation was increased at 1 h after PEDV infection, in accordance with the expression of TSPAN3, CD63, and ITGB2. Similar trends appeared in the intestines of piglets in the early stage of PEDV challenge. Compared with Vero cells, S209A-Vero cells in which eIF4E cannot be phosphorylated showed a decrease of invading PEDV virions. MNK kinase inhibitor blocked PEDV invasion, as well as reduced the accumulation of TSPAN3, CD63, and ITGB2. Further study showed that the ERK-MNK pathway was responsible for the regulation of PEDV-induced early phosphorylation of eIF4E. This paper demonstrates for the first time the connections among p-eIF4E stimulation and membrane-residential host factors. Our findings also enrich the understanding of the biological function of phosphorylated eIF4E during the viral life cycle.IMPORTANCEThe eukaryotic translation initiation factor eIF4E can regulate cellular translation via phosphorylation. In our previous study, several host factors susceptible to a high level of p-eIF4E were found to be conducive to viral infection by coronavirus PEDV. The current paper is focused on cell membrane-residential factors, which are involved in signal pathways that are sensitive to phosphorylated eIF4E. We found that the ERK-MNK pathway was activated, which resulted in the stimulation of phosphorylation of eIF4E in early PEDV infection. Phospho-eIF4E promoted the viral invasion of PEDV by upregulating the expression of host factors TSPAN3, CD63, and ITGB2 at the translation level rather than at the transcription level. Moreover, TSPAN3, CD63, or ITGB2 facilitates the efficient entry of coronavirus SARS-CoV, SARS-CoV-2, and HCoV-OC43. Our findings broaden our insights into the dynamic phosphorylation of eIF4E during the viral life cycle, and provide further evidence that phosphorylated eIF4E regulates selective translation of host mRNA.
Collapse
Affiliation(s)
- Xiu-Zhong Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wen-Jun Tian
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing-Ling You
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiao-Jing Chi
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao-Jia Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Cai Z, Bai H, Ren D, Xue B, Liu Y, Gong T, Zhang X, Zhang P, Zhu J, Shi B, Zhang C. Integrin αvβ1 facilitates ACE2-mediated entry of SARS-CoV-2. Virus Res 2024; 339:199251. [PMID: 37884208 PMCID: PMC10651773 DOI: 10.1016/j.virusres.2023.199251] [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: 03/30/2023] [Revised: 06/14/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Integrins have been suggested to be involved in SARS-CoV-2 infection, but the underlying mechanisms remain largely unclear. This study aimed to investigate how integrins facilitate the ACE2-mediated cellular entry of SARS-CoV-2. We first tested the susceptibility of a panel of human cell lines to SARS-CoV-2 infection using the spike protein pseudotyped virus assay and examined the expression levels of integrins in these cell lines by qPCR, western blot and flow cytometry. We found that integrin αvβ1 was highly enriched in the SARS-CoV-2 susceptible cell lines. Additional studies demonstrated that RGD (403-405)→AAA mutant was defective in binding to integrin αvβ1 compared to its wild type counterpart, and anti-αvβ1 integrin antibodies significantly inhibited the entry of SARS-CoV-2 into the cells. Further studies using mouse NIH3T3 cells expressing human ACE2, integrin αv, integrin β1, and/or integrin αvβ1 suggest that integrin αvβ1 was unable to function as an independent receptor but could significantly facilitate the cellular entry of SASR-CoV-2. Finally, we observed that the Omicron exhibited a significant increase in the ACE2-mediated viral entry. Our findings may enhance our understanding of the pathogenesis of SARS-CoV-2 infection and offer potential therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Zeqiong Cai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Doudou Ren
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Biyun Xue
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Yijia Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Tian Gong
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China; Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China
| | - Xuan Zhang
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China; Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China
| | - Peng Zhang
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China; Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China
| | - Junsheng Zhu
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Binyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China.
| | - Chengsheng Zhang
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China; Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China; Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie, Nanchang 330006, China.
| |
Collapse
|
8
|
Zhang H, Wang Z, Nguyen HTT, Watson AJ, Lao Q, Li A, Zhu J. Integrin α 5β 1 contributes to cell fusion and inflammation mediated by SARS-CoV-2 spike via RGD-independent interaction. Proc Natl Acad Sci U S A 2023; 120:e2311913120. [PMID: 38060559 PMCID: PMC10723138 DOI: 10.1073/pnas.2311913120] [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: 07/17/2023] [Accepted: 11/03/2023] [Indexed: 12/17/2023] Open
Abstract
The Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infects host cells by engaging its spike (S) protein with human ACE2 receptor. Recent studies suggest the involvement of integrins in SARS-CoV-2 infection through interaction with the S protein, but the underlying mechanism is not well understood. This study investigated the role of integrin α5β1, which recognizes the Arg-Gly-Asp (RGD) motif in its physiological ligands, in S-mediated virus entry and cell-cell fusion. Our results showed that α5β1 does not directly contribute to S-mediated cell entry, but it enhances S-mediated cell-cell fusion in collaboration with ACE2. This effect cannot be inhibited by the putative α5β1 inhibitor ATN-161 or the high-affinity RGD-mimetic inhibitor MK-0429 but requires the participation of α5 cytoplasmic tail (CT). We detected a direct interaction between α5β1 and the S protein, but this interaction does not rely on the RGD-containing receptor binding domain of the S1 subunit of the S protein. Instead, it involves the S2 subunit of the S protein and α5β1 homo-oligomerization. Furthermore, we found that the S protein induces inflammatory responses in human endothelial cells, characterized by NF-κB activation, gasdermin D cleavage, and increased secretion of proinflammatory cytokines IL-6 and IL-1β. These effects can be attenuated by the loss of α5 expression or inhibition of the α5 CT binding protein phosphodiesterase-4D (PDE4D), suggesting the involvement of α5 CT and PDE4D pathway. These findings provide molecular insights into the pathogenesis of SARS-CoV-2 mediated by a nonclassical RGD-independent ligand-binding and signaling function of integrin α5β1 and suggest potential targets for antiviral treatment.
Collapse
Affiliation(s)
- Heng Zhang
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - Zhengli Wang
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - Huong T. T. Nguyen
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - Abigail J. Watson
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - Qifang Lao
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - An Li
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
| | - Jieqing Zhu
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI53226
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI53226
| |
Collapse
|
9
|
Fiori F, Cossu FL, Salis F, Carboni D, Stagi L, De Forni D, Poddesu B, Malfatti L, Khalel A, Salis A, Casula MF, Anedda R, Lori F, Innocenzi P. In Vitro Antiviral Activity of Hyperbranched Poly-L-Lysine Modified by L-Arginine against Different SARS-CoV-2 Variants. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3090. [PMID: 38132987 PMCID: PMC10745586 DOI: 10.3390/nano13243090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
The emergence of SARS-CoV-2 variants requires close monitoring to prevent the reoccurrence of a new pandemic in the near future. The Omicron variant, in particular, is one of the fastest-spreading viruses, showing a high ability to infect people and evade neutralization by antibodies elicited upon infection or vaccination. Therefore, the search for broad-spectrum antivirals that can inhibit the infectious capacity of SARS-CoV-2 is still the focus of intense research. In the present work, hyperbranched poly-L-lysine nanopolymers, which have shown an excellent ability to block the original strain of SARS-CoV-2 infection, were modified with L-arginine. A thermal reaction at 240 °C catalyzed by boric acid yielded Lys-Arg hyperbranched nanopolymers. The ability of these nanopolymers to inhibit viral replication were assessed for the original, Delta, and Omicron strains of SARS-CoV-2 together with their cytotoxicity. A reliable indication of the safety profile and effectiveness of the various polymeric compositions in inhibiting or suppressing viral infection was obtained by the evaluation of the therapeutic index in an in vitro prevention model. The hyperbranched L-arginine-modified nanopolymers exhibited a twelve-fold greater therapeutic index when tested with the original strain. The nanopolymers could also effectively limit the replication of the Omicron strain in a cell culture.
Collapse
Affiliation(s)
- Federico Fiori
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Franca Lucia Cossu
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Federica Salis
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Davide Carboni
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Luigi Stagi
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Davide De Forni
- ViroStatics srl, Viale Umberto I, 46, 07100 Sassari, Italy; (D.D.F.); (B.P.); (F.L.)
| | - Barbara Poddesu
- ViroStatics srl, Viale Umberto I, 46, 07100 Sassari, Italy; (D.D.F.); (B.P.); (F.L.)
| | - Luca Malfatti
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| | - Abbas Khalel
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Andrea Salis
- Department of Chemical and Geolocial Sciences, University of Cagliari, Cittadella Universitaria SS 554 Bivio Sestu, 09042 Monserrato, Italy;
| | - Maria Francesca Casula
- Department of Mechanical, Chemical and Materials Engineering, University of Cagliari, Via Marengo, 2, 09123 Cagliari, Italy
| | - Roberto Anedda
- Porto Conte Ricerche srl, Strada Provinciale S.P. 55, Loc. Tramariglio, 07041 Alghero, Italy;
| | - Franco Lori
- ViroStatics srl, Viale Umberto I, 46, 07100 Sassari, Italy; (D.D.F.); (B.P.); (F.L.)
| | - Plinio Innocenzi
- Laboratory of Materials Science and Nanotechnology (LMNT), CR-INSTM, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.F.); (F.L.C.); (F.S.); (D.C.); (L.S.); (L.M.)
| |
Collapse
|
10
|
Sha A, Liu Y, Hao H. Current state-of-the-art and potential future therapeutic drugs against COVID-19. Front Cell Dev Biol 2023; 11:1238027. [PMID: 37691829 PMCID: PMC10485263 DOI: 10.3389/fcell.2023.1238027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
The novel coronavirus disease (COVID-19) continues to endanger human health, and its therapeutic drugs are under intensive research and development. Identifying the efficacy and toxicity of drugs in animal models is helpful for further screening of effective medications, which is also a prerequisite for drugs to enter clinical trials. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) invades host cells mainly by the S protein on its surface. After the SARS-CoV-2 RNA genome is injected into the cells, M protein will help assemble and release new viruses. RdRp is crucial for virus replication, assembly, and release of new virus particles. This review analyzes and discusses 26 anti-SARS-CoV-2 drugs based on their mechanism of action, effectiveness and safety in different animal models. We propose five drugs to be the most promising to enter the next stage of clinical trial research, thus providing a reference for future drug development.
Collapse
Affiliation(s)
- Ailong Sha
- School of Teacher Education, Chongqing Three Gorges University, Chongqing, China
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Yi Liu
- School of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Haiyan Hao
- School of Environmental and Chemical Engineering, Chongqing, China
| |
Collapse
|
11
|
Sacchi A, Giannessi F, Sabatini A, Percario ZA, Affabris E. SARS-CoV-2 Evasion of the Interferon System: Can We Restore Its Effectiveness? Int J Mol Sci 2023; 24:ijms24119353. [PMID: 37298304 DOI: 10.3390/ijms24119353] [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: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Type I and III Interferons (IFNs) are the first lines of defense in microbial infections. They critically block early animal virus infection, replication, spread, and tropism to promote the adaptive immune response. Type I IFNs induce a systemic response that impacts nearly every cell in the host, while type III IFNs' susceptibility is restricted to anatomic barriers and selected immune cells. Both IFN types are critical cytokines for the antiviral response against epithelium-tropic viruses being effectors of innate immunity and regulators of the development of the adaptive immune response. Indeed, the innate antiviral immune response is essential to limit virus replication at the early stages of infection, thus reducing viral spread and pathogenesis. However, many animal viruses have evolved strategies to evade the antiviral immune response. The Coronaviridae are viruses with the largest genome among the RNA viruses. Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) pandemic. The virus has evolved numerous strategies to contrast the IFN system immunity. We intend to describe the virus-mediated evasion of the IFN responses by going through the main phases: First, the molecular mechanisms involved; second, the role of the genetic background of IFN production during SARS-CoV-2 infection; and third, the potential novel approaches to contrast viral pathogenesis by restoring endogenous type I and III IFNs production and sensitivity at the sites of infection.
Collapse
Affiliation(s)
- Alessandra Sacchi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Flavia Giannessi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Andrea Sabatini
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Zulema Antonia Percario
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Elisabetta Affabris
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| |
Collapse
|
12
|
Iaconis D, Caccuri F, Manelfi C, Talarico C, Bugatti A, Filippini F, Zani A, Novelli R, Kuzikov M, Ellinger B, Gribbon P, Riecken K, Esposito F, Corona A, Tramontano E, Beccari AR, Caruso A, Allegretti M. DHFR Inhibitors Display a Pleiotropic Anti-Viral Activity against SARS-CoV-2: Insights into the Mechanisms of Action. Viruses 2023; 15:v15051128. [PMID: 37243214 DOI: 10.3390/v15051128] [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: 04/05/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
During the COVID-19 pandemic, drug repurposing represented an effective strategy to obtain quick answers to medical emergencies. Based on previous data on methotrexate (MTX), we evaluated the anti-viral activity of several DHFR inhibitors in two cell lines. We observed that this class of compounds showed a significant influence on the virus-induced cytopathic effect (CPE) partly attributed to the intrinsic anti-metabolic activity of these drugs, but also to a specific anti-viral function. To elucidate the molecular mechanisms, we took advantage of our EXSCALATE platform for in-silico molecular modelling and further validated the influence of these inhibitors on nsp13 and viral entry. Interestingly, pralatrexate and trimetrexate showed superior effects in counteracting the viral infection compared to other DHFR inhibitors. Our results indicate that their higher activity is due to their polypharmacological and pleiotropic profile. These compounds can thus potentially give a clinical advantage in the management of SARS-CoV-2 infection in patients already treated with this class of drugs.
Collapse
Affiliation(s)
- Daniela Iaconis
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131 Napoli, Italy
| | - Francesca Caccuri
- Section of Microbiology Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Candida Manelfi
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131 Napoli, Italy
| | - Carmine Talarico
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131 Napoli, Italy
| | - Antonella Bugatti
- Section of Microbiology Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Federica Filippini
- Section of Microbiology Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alberto Zani
- Section of Microbiology Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Rubina Novelli
- Dompè Famaceutici SpA, Via Campo di Pile snc, 67100 L'Aquila, Italy
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria SS554, 09042 Monserrato (CA), Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria SS554, 09042 Monserrato (CA), Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria SS554, 09042 Monserrato (CA), Italy
| | | | - Arnaldo Caruso
- Section of Microbiology Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | | |
Collapse
|
13
|
Ma X, Liang J, Zhu G, Bhoria P, Shoara AA, MacKeigan DT, Khoury CJ, Slavkovic S, Lin L, Karakas D, Chen Z, Prifti V, Liu Z, Shen C, Li Y, Zhang C, Dou J, Rousseau Z, Zhang J, Ni T, Lei X, Chen P, Wu X, Shaykhalishahi H, Mubareka S, Connelly KA, Zhang H, Rotstein O, Ni H. SARS-CoV-2 RBD and Its Variants Can Induce Platelet Activation and Clearance: Implications for Antibody Therapy and Vaccinations against COVID-19. RESEARCH (WASHINGTON, D.C.) 2023; 6:0124. [PMID: 37223472 PMCID: PMC10202384 DOI: 10.34133/research.0124] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/28/2023] [Indexed: 10/10/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 virus is an ongoing global health burden. Severe cases of COVID-19 and the rare cases of COVID-19 vaccine-induced-thrombotic-thrombocytopenia (VITT) are both associated with thrombosis and thrombocytopenia; however, the underlying mechanisms remain inadequately understood. Both infection and vaccination utilize the spike protein receptor-binding domain (RBD) of SARS-CoV-2. We found that intravenous injection of recombinant RBD caused significant platelet clearance in mice. Further investigation revealed the RBD could bind platelets, cause platelet activation, and potentiate platelet aggregation, which was exacerbated in the Delta and Kappa variants. The RBD-platelet interaction was partially dependent on the β3 integrin as binding was significantly reduced in β3-/- mice. Furthermore, RBD binding to human and mouse platelets was significantly reduced with related αIIbβ3 antagonists and mutation of the RGD (arginine-glycine-aspartate) integrin binding motif to RGE (arginine-glycine-glutamate). We developed anti-RBD polyclonal and several monoclonal antibodies (mAbs) and identified 4F2 and 4H12 for their potent dual inhibition of RBD-induced platelet activation, aggregation, and clearance in vivo, and SARS-CoV-2 infection and replication in Vero E6 cells. Our data show that the RBD can bind platelets partially though αIIbβ3 and induce platelet activation and clearance, which may contribute to thrombosis and thrombocytopenia observed in COVID-19 and VITT. Our newly developed mAbs 4F2 and 4H12 have potential not only for diagnosis of SARS-CoV-2 virus antigen but also importantly for therapy against COVID-19.
Collapse
Affiliation(s)
- Xiaoying Ma
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Jady Liang
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Department of Physiology,
University of Toronto, Toronto, ON, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
| | - Aron A. Shoara
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Department of Physiology,
University of Toronto, Toronto, ON, Canada
| | - Daniel T. MacKeigan
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Department of Physiology,
University of Toronto, Toronto, ON, Canada
| | - Christopher J. Khoury
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Sladjana Slavkovic
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
| | - Lisha Lin
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Danielle Karakas
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Ziyan Chen
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
| | - Viktor Prifti
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Zhenze Liu
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Yuchong Li
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease,
The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Cheng Zhang
- CCOA Therapeutics Inc., Toronto, ON, Canada
- Department of Laboratory Medicine,
The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiayu Dou
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Zack Rousseau
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Jiamin Zhang
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Tiffany Ni
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
| | - Xi Lei
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
| | - Pingguo Chen
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
| | - Xiaoyu Wu
- Advanced Pharmaceutics & Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, Toronto, ON, Canada
| | - Hamed Shaykhalishahi
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
| | - Samira Mubareka
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Medical Microbiology and Infectious Disease,
Sunnybrook Health Science Centre, Toronto, ON, Canada
| | - Kim A. Connelly
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Department of Medicine,
University of Toronto, Toronto, ON, Canada
- Division of Cardiology,
St. Michael's Hospital, Toronto, ON, Canada
| | - Haibo Zhang
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease,
The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Medical Microbiology and Infectious Disease,
Sunnybrook Health Science Centre, Toronto, ON, Canada
- Department of Anesthesiology and Pain Medicine and Division of Critical Care Medicine,
University of Toronto, Toronto, ON, Canada
- Interdepartmental Division of Critical Care Medicine,
University of Toronto, Toronto, ON, Canada
| | - Ori Rotstein
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery,
University of Toronto, Toronto, ON, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine,
Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Department of Physiology,
University of Toronto, Toronto, ON, Canada
- CCOA Therapeutics Inc., Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
- Department of Medicine,
University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Petito E, Franco L, Falcinelli E, Guglielmini G, Conti C, Vaudo G, Paliani U, Becattini C, Mencacci A, Tondi F, Gresele P. COVID-19 infection-associated platelet and neutrophil activation is blunted by previous anti-SARS-CoV-2 vaccination. Br J Haematol 2023; 201:851-856. [PMID: 36883298 DOI: 10.1111/bjh.18726] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 03/09/2023]
Abstract
The effectiveness of vaccination against SARS-CoV-2 in preventing COVID-19 or in reducing severe illness in subjects hospitalized for COVID-19 despite vaccination has been unequivocally shown. However, no studies so far have assessed if subjects who get COVID-19 despite vaccination are protected from SARS-CoV-2-induced platelet, neutrophil and endothelial activation, biomarkers associated with thrombosis and worse outcome. In this pilot study, we show that previous vaccination blunts COVID-19-associated platelet activation, assessed by circulating platelet-derived microvesicles and soluble P-selectin, and neutrophil activation, assessed by circulating neutrophil extracellular trap (NET) biomarkers and matrix metalloproteinase-9, and reduces COVID-19-associated thrombotic events, hospitalization in intensive-care units and death.
Collapse
Affiliation(s)
- Eleonora Petito
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Laura Franco
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Emanuela Falcinelli
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Guglielmini
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Chiara Conti
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Gaetano Vaudo
- Unit of Internal Medicine, Terni University Hospital, Terni, Italy
| | - Ugo Paliani
- Division of Internal Medicine, USL Umbria 1, Pantalla, Italy
| | - Cecilia Becattini
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Antonella Mencacci
- Department of Medicine and Surgery, Microbiology and Clinical Microbiology, University of Perugia, Perugia, Italy
| | - Francesca Tondi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
15
|
Matsumoto C, Shibata S, Kishi T, Morimoto S, Mogi M, Yamamoto K, Kobayashi K, Tanaka M, Asayama K, Yamamoto E, Nakagami H, Hoshide S, Mukoyama M, Kario K, Node K, Rakugi H. Long COVID and hypertension-related disorders: a report from the Japanese Society of Hypertension Project Team on COVID-19. Hypertens Res 2023; 46:601-619. [PMID: 36575228 PMCID: PMC9793823 DOI: 10.1038/s41440-022-01145-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
The coronavirus disease 2019 (COVID-19) affects infected patients even after the acute phase and impairs their health and quality of life by causing a wide variety of symptoms, referred to as long COVID. Although the evidence is still insufficient, hypertension is suspected to be a potential risk factor for long COVID, and the occurrence of cardiovascular diseases seems to be a key facet of multiple conditions observed in long COVID. Nonetheless, there are few reports that comprehensively review the impacts of long COVID on hypertension and related disorders. As a sequel to our previous report in 2020 which reviewed the association of COVID-19 and hypertension, we summarize the possible influences of long COVID on hypertension-related organs, including the cardiovascular system, kidney, and endocrine system, as well as the pathophysiological mechanisms associated with the disorders in this review. Given that the clinical course of COVID-19 is highly affected by age and sex, we also review the impacts of these factors on long COVID. Lastly, we discuss areas of uncertainty and future directions, which may lead to better understanding and improved prognosis of clinical problems associated with COVID-19.
Collapse
Affiliation(s)
- Chisa Matsumoto
- Department of Cardiology, Preventive medicine, Tokyo Medical University, Tokyo, Japan.
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.
| | - Takuya Kishi
- Department of Graduate School of Medicine (Cardiology), International University of Health and Welfare, Okawa, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuo Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Kobayashi Internal Medicine Clinic, Sagamihara, Japan
| | - Masami Tanaka
- Department of Internal Medicine, Adachi Medical Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Kei Asayama
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Satoshi Hoshide
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
16
|
Norris EG, Pan XS, Hocking DC. Receptor-binding domain of SARS-CoV-2 is a functional αv-integrin agonist. J Biol Chem 2023; 299:102922. [PMID: 36669646 PMCID: PMC9846890 DOI: 10.1016/j.jbc.2023.102922] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Among the novel mutations distinguishing SARS-CoV-2 from similar coronaviruses is a K403R substitution in the receptor-binding domain (RBD) of the viral spike (S) protein within its S1 region. This amino acid substitution occurs near the angiotensin-converting enzyme 2-binding interface and gives rise to a canonical RGD adhesion motif that is often found in native extracellular matrix proteins, including fibronectin. Here, the ability of recombinant S1-RBD to bind to cell surface integrins and trigger downstream signaling pathways was assessed and compared with RGD-containing, integrin-binding fragments of fibronectin. We determined that S1-RBD supported adhesion of fibronectin-null mouse embryonic fibroblasts as well as primary human small airway epithelial cells, while RBD-coated microparticles attached to epithelial monolayers in a cation-dependent manner. Cell adhesion to S1-RBD was RGD dependent and inhibited by blocking antibodies against αv and β3 but not α5 or β1 integrins. Similarly, we observed direct binding of S1-RBD to recombinant human αvβ3 and αvβ6 integrins, but not α5β1 integrins, using surface plasmon resonance. S1-RBD adhesion initiated cell spreading, focal adhesion formation, and actin stress fiber organization to a similar extent as fibronectin. Moreover, S1-RBD stimulated tyrosine phosphorylation of the adhesion mediators FAK, Src, and paxillin; triggered Akt activation; and supported cell proliferation. Thus, the RGD sequence of S1-RBD can function as an αv-selective integrin agonist. This study provides evidence that cell surface αv-containing integrins can respond functionally to spike protein and raises the possibility that S1-mediated dysregulation of extracellular matrix dynamics may contribute to the pathogenesis and/or post-acute sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emma G Norris
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Xuan Sabrina Pan
- Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Denise C Hocking
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
17
|
Bellavite P, Ferraresi A, Isidoro C. Immune Response and Molecular Mechanisms of Cardiovascular Adverse Effects of Spike Proteins from SARS-CoV-2 and mRNA Vaccines. Biomedicines 2023; 11:451. [PMID: 36830987 PMCID: PMC9953067 DOI: 10.3390/biomedicines11020451] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
The SARS-CoV-2 (severe acute respiratory syndrome coronavirus responsible for the COVID-19 disease) uses the Spike proteins of its envelope for infecting target cells expressing on the membrane the angiotensin converting enzyme 2 (ACE2) enzyme that acts as a receptor. To control the pandemic, genetically engineered vaccines have been designed for inducing neutralizing antibodies against the Spike proteins. These vaccines do not act like traditional protein-based vaccines, as they deliver the message in the form of mRNA or DNA to host cells that then produce and expose the Spike protein on the membrane (from which it can be shed in soluble form) to alert the immune system. Mass vaccination has brought to light various adverse effects associated with these genetically based vaccines, mainly affecting the circulatory and cardiovascular system. ACE2 is present as membrane-bound on several cell types, including the mucosa of the upper respiratory and of the gastrointestinal tracts, the endothelium, the platelets, and in soluble form in the plasma. The ACE2 enzyme converts the vasoconstrictor angiotensin II into peptides with vasodilator properties. Here we review the pathways for immunization and the molecular mechanisms through which the Spike protein, either from SARS-CoV-2 or encoded by the mRNA-based vaccines, interferes with the Renin-Angiotensin-System governed by ACE2, thus altering the homeostasis of the circulation and of the cardiovascular system. Understanding the molecular interactions of the Spike protein with ACE2 and the consequent impact on cardiovascular system homeostasis will direct the diagnosis and therapy of the vaccine-related adverse effects and provide information for development of a personalized vaccination that considers pathophysiological conditions predisposing to such adverse events.
Collapse
Affiliation(s)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
18
|
Bugatti A, Filippini F, Messali S, Giovanetti M, Ravelli C, Zani A, Ciccozzi M, Caruso A, Caccuri F. The D405N Mutation in the Spike Protein of SARS-CoV-2 Omicron BA.5 Inhibits Spike/Integrins Interaction and Viral Infection of Human Lung Microvascular Endothelial Cells. Viruses 2023; 15:332. [PMID: 36851546 PMCID: PMC9962894 DOI: 10.3390/v15020332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
Severe COVID-19 is characterized by angiogenic features, such as intussusceptive angiogenesis, endothelialitis, and activation of procoagulant pathways. This pathological state can be ascribed to a direct SARS-CoV-2 infection of human lung ECs. Recently, we showed the capability of SARS-CoV-2 to infect ACE2-negative primary human lung microvascular endothelial cells (HL-mECs). This occurred through the interaction of an Arg-Gly-Asp (RGD) motif, endowed on the Spike protein at position 403-405, with αvβ3 integrin expressed on HL-mECs. HL-mEC infection promoted the remodeling of cells toward a pro-inflammatory and pro-angiogenic phenotype. The RGD motif is distinctive of SARS-CoV-2 Spike proteins up to the Omicron BA.1 subvariant. Suddenly, a dominant D405N mutation was expressed on the Spike of the most recently emerged Omicron BA.2, BA.4, and BA.5 subvariants. Here we demonstrate that the D405N mutation inhibits Omicron BA.5 infection of HL-mECs and their dysfunction because of the lack of Spike/integrins interaction. The key role of ECs in SARS-CoV-2 pathogenesis has been definitively proven. Evidence of mutations retrieving the capability of SARS-CoV-2 to infect HL-mECs highlights a new scenario for patients infected with the newly emerged SARS-CoV-2 Omicron subvariants, suggesting that they may display less severe disease manifestations than those observed with previous variants.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Federica Filippini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Marta Giovanetti
- Laboratório de Flavivirus, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
- Department of Science and Technology for Humans and the Environment, University Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Cosetta Ravelli
- Section of General Pathology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
19
|
Fajloun Z, Legros C, Sabatier JM. COVID-19 and Ehlers-Danlos Syndrome: The Dangers of the Spike Protein of SARS-CoV-2. Infect Disord Drug Targets 2023; 23:e040123212375. [PMID: 36600622 DOI: 10.2174/1871526523666230104145108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Affiliation(s)
- Ziad Fajloun
- Department of Biology, Faculty of Sciences 3, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Christian Legros
- College of Life Sciences, University of Angers, INSERM, CNRS, MITOVASC, Team 2 CarMe, SFR ICAT, Angers 49000, France
| | - Jean-Marc Sabatier
- College of Life Sciences, Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, Marseille 13385, France
| |
Collapse
|
20
|
Griffin KH, Fok SW, Kent Leach J. Strategies to capitalize on cell spheroid therapeutic potential for tissue repair and disease modeling. NPJ Regen Med 2022; 7:70. [PMID: 36494368 PMCID: PMC9734656 DOI: 10.1038/s41536-022-00266-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Cell therapies offer a tailorable, personalized treatment for use in tissue engineering to address defects arising from trauma, inefficient wound repair, or congenital malformation. However, most cell therapies have achieved limited success to date. Typically injected in solution as monodispersed cells, transplanted cells exhibit rapid cell death or insufficient retention at the site, thereby limiting their intended effects to only a few days. Spheroids, which are dense, three-dimensional (3D) aggregates of cells, enhance the beneficial effects of cell therapies by increasing and prolonging cell-cell and cell-matrix signaling. The use of spheroids is currently under investigation for many cell types. Among cells under evaluation, spheroids formed of mesenchymal stromal cells (MSCs) are particularly promising. MSC spheroids not only exhibit increased cell survival and retained differentiation, but they also secrete a potent secretome that promotes angiogenesis, reduces inflammation, and attracts endogenous host cells to promote tissue regeneration and repair. However, the clinical translation of spheroids has lagged behind promising preclinical outcomes due to hurdles in their formation, instruction, and use that have yet to be overcome. This review will describe the current state of preclinical spheroid research and highlight two key examples of spheroid use in clinically relevant disease modeling. It will highlight techniques used to instruct the phenotype and function of spheroids, describe current limitations to their use, and offer suggestions for the effective translation of cell spheroids for therapeutic treatments.
Collapse
Affiliation(s)
- Katherine H Griffin
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
- School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Shierly W Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
21
|
Pelisek J, Reutersberg B, Greber UF, Zimmermann A. Vascular dysfunction in COVID-19 patients: update on SARS-CoV-2 infection of endothelial cells and the role of long non-coding RNAs. Clin Sci (Lond) 2022; 136:1571-1590. [PMID: 36367091 PMCID: PMC9652506 DOI: 10.1042/cs20220235] [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: 04/08/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 08/16/2023]
Abstract
Although COVID-19 is primarily a respiratory disease, it may affect also the cardiovascular system. COVID-19 patients with cardiovascular disorder (CVD) develop a more severe disease course with a significantly higher mortality rate than non-CVD patients. A common denominator of CVD is the dysfunction of endothelial cells (ECs), increased vascular permeability, endothelial-to-mesenchymal transition, coagulation, and inflammation. It has been assumed that clinical complications in COVID-19 patients suffering from CVD are caused by SARS-CoV-2 infection of ECs through the angiotensin-converting enzyme 2 (ACE2) receptor and the cellular transmembrane protease serine 2 (TMPRSS2) and the consequent dysfunction of the infected vascular cells. Meanwhile, other factors associated with SARS-CoV-2 entry into the host cells have been described, including disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), the C-type lectin CD209L or heparan sulfate proteoglycans (HSPG). Here, we discuss the current data about the putative entry of SARS-CoV-2 into endothelial and smooth muscle cells. Furthermore, we highlight the potential role of long non-coding RNAs (lncRNAs) affecting vascular permeability in CVD, a process that might exacerbate disease in COVID-19 patients.
Collapse
Affiliation(s)
- Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zürich, Zürich, Switzerland
| | | | - Urs F Greber
- Department of Molecular Life Sciences, University of Zürich, Switzerland
| | | |
Collapse
|
22
|
De Forni D, Poddesu B, Cugia G, Chafouleas J, Lisziewicz J, Lori F. Synergistic drug combinations designed to fully suppress SARS-CoV-2 in the lung of COVID-19 patients. PLoS One 2022; 17:e0276751. [PMID: 36355808 PMCID: PMC9648746 DOI: 10.1371/journal.pone.0276751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/12/2022] [Indexed: 11/12/2022] Open
Abstract
Despite new antivirals are being approved against SARS-CoV-2 they suffer from significant constraints and are not indicated for hospitalized patients, who are left with few antiviral options. Repurposed drugs have previously shown controversial clinical results and it remains difficult to understand why certain trials delivered positive results and other trials failed. Our manuscript contributes to explaining the puzzle: this might have been caused by a suboptimal drug exposure and, consequently, an incomplete virus suppression, also because the drugs have mostly been used as add-on monotherapies. As with other viruses (e.g., HIV and HCV) identifying synergistic combinations among such drugs could overcome monotherapy-related limitations. In a cell culture model for SARS-CoV-2 infection the following stringent criteria were adopted to assess drug combinations: 1) identify robust, synergistic antiviral activity with no increase in cytotoxicity, 2) identify the lowest drug concentration inhibiting the virus by 100% (LIC100) and 3) understand whether the LIC100 could be reached in the lung at clinically indicated drug doses. Among several combinations tested, remdesivir with either azithromycin or ivermectin synergistically increased the antiviral activity with no increase in cytotoxicity, improving the therapeutic index and lowering the LIC100 of every one of the drugs to levels that are expected to be achievable and maintained in the lung for a therapeutically relevant period of time. These results are consistent with recent clinical observations showing that intensive care unit admission was significantly delayed by the combination of AZI and RDV, but not by RDV alone, and could have immediate implications for the treatment of hospitalized patients with COVID-19 as the proposed "drug cocktails" should have antiviral activity against present and future SARS-CoV-2 variants without significant overlapping toxicity, while minimizing the onset of drug resistance. Our results also provide a validated methodology to help sort out which combination of drugs are most likely to be efficacious in vivo, based on their in vitro activity, potential synergy and PK profiles.
Collapse
Affiliation(s)
| | | | | | | | - Julianna Lisziewicz
- Research Institute for Genetic and Human Therapy, Colorado Springs, CO, United States of America
| | - Franco Lori
- ViroStatics s.r.l., Tramariglio, Italy
- Research Institute for Genetic and Human Therapy, Colorado Springs, CO, United States of America
| |
Collapse
|
23
|
de Melo TC, Trevisan-Silva D, Alvarez-Flores MP, Gomes RN, de Souza MM, Valerio HP, Oliveira DS, DeOcesano-Pereira C, Botosso VF, Calil Jorge SA, Schattner M, Gomez RM, Chudzinski-Tavassi AM. Proteomic Analysis Identifies Molecular Players and Biological Processes Specific to SARS-CoV-2 Exposure in Endothelial Cells. Int J Mol Sci 2022; 23:10452. [PMID: 36142365 PMCID: PMC9500950 DOI: 10.3390/ijms231810452] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been responsible for the severe pandemic of acute respiratory disease, coronavirus disease 2019 (COVID-19), experienced in the 21st century. The clinical manifestations range from mild symptoms to abnormal blood coagulation and severe respiratory failure. In severe cases, COVID-19 manifests as a thromboinflammatory disease. Damage to the vascular compartment caused by SARS-CoV-2 has been linked to thrombosis, triggered by an enhanced immune response. The molecular mechanisms underlying endothelial activation have not been fully elucidated. We aimed to identify the proteins correlated to the molecular response of human umbilical vein endothelial cells (HUVECs) after exposure to SARS-CoV-2, which might help to unravel the molecular mechanisms of endothelium activation in COVID-19. In this direction, we exposed HUVECs to SARS-CoV-2 and analyzed the expression of specific cellular receptors, and changes in the proteome of HUVECs at different time points. We identified that HUVECs exhibit non-productive infection without cytopathic effects, in addition to the lack of expression of specific cell receptors known to be essential for SARS-CoV-2 entry into cells. We highlighted the enrichment of the protein SUMOylation pathway and the increase in SUMO2, which was confirmed by orthogonal assays. In conclusion, proteomic analysis revealed that the exposure to SARS-CoV-2 induced oxidative stress and changes in protein abundance and pathways enrichment that resembled endothelial dysfunction.
Collapse
Affiliation(s)
- Thatiana Corrêa de Melo
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Dilza Trevisan-Silva
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Miryam P. Alvarez-Flores
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Renata Nascimento Gomes
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Marcelo Medina de Souza
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Hellen Paula Valerio
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Douglas S. Oliveira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | | | | | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX-CONICET-ANM), Buenos Aires 1425, Argentina
| | - Ricardo M. Gomez
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, CONICET-UNLP, La Plata 1900, Argentina
| | | |
Collapse
|
24
|
Yan K, Dumenil T, Tang B, Le TT, Bishop CR, Suhrbier A, Rawle DJ. Evolution of ACE2-independent SARS-CoV-2 infection and mouse adaption after passage in cells expressing human and mouse ACE2. Virus Evol 2022; 8:veac063. [PMID: 35919871 PMCID: PMC9338707 DOI: 10.1093/ve/veac063] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022] Open
Abstract
Human ACE2 Human angiotensin converting enzyme 2 (hACE2) is the key cell attachment and entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with the original SARS-CoV-2 isolates unable to use mouse ACE2 (mACE2). Herein we describe the emergence of a SARS-CoV-2 strain capable of ACE2-independent infection and the evolution of mouse-adapted (MA) SARS-CoV-2 by in vitro serial passaging of virus in co-cultures of cell lines expressing hACE2 and mACE2. MA viruses evolved with up to five amino acid changes in the spike protein, all of which have been seen in human isolates. MA viruses replicated to high titers in C57BL/6J mouse lungs and nasal turbinates and caused characteristic lung histopathology. One MA virus also evolved to replicate efficiently in several ACE2-negative cell lines across several species, including clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) ACE2 knockout cells. An E484D substitution is likely involved in ACE2-independent entry and has appeared in only ≈0.003 per cent of human isolates globally, suggesting that it provided no significant selection advantage in humans. ACE2-independent entry reveals a SARS-CoV-2 infection mechanism that has potential implications for disease pathogenesis, evolution, tropism, and perhaps also intervention development.
Collapse
Affiliation(s)
- Kexin Yan
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| | - Troy Dumenil
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| | - Bing Tang
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| | - Thuy T Le
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| | - Cameron R Bishop
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| | - Andreas Suhrbier
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, 300 Herston Road, Herston, 4029 and The University of Queensland, St Lucia, 4072, Australia
| | - Daniel J Rawle
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4029, Queensland, Australia
| |
Collapse
|
25
|
Norris EG, Pan XS, Hocking DC. Receptor binding domain of SARS-CoV-2 is a functional αv-integrin agonist. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.04.11.487882. [PMID: 35441172 PMCID: PMC9016641 DOI: 10.1101/2022.04.11.487882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Among the novel mutations distinguishing SARS-CoV-2 from similar respiratory coronaviruses is a K403R substitution in the receptor-binding domain (RBD) of the viral spike (S) protein within its S1 region. This amino acid substitution occurs near the angiotensin-converting enzyme 2 (ACE2)-binding interface and gives rise to a canonical RGD adhesion motif that is often found in native extracellular matrix proteins, including fibronectin. In the present study, the ability of recombinant S1-RBD to bind to cell surface integrins and trigger downstream signaling pathways was assessed and compared to RGD-containing, integrin-binding fragments of fibronectin. S1-RBD supported adhesion of both fibronectin-null mouse embryonic fibroblasts as well as primary human small airway epithelial cells. Cell adhesion to S1-RBD was cation- and RGD-dependent, and was inhibited by blocking antibodies against α v and β 3 , but not α 5 or β 1 , integrins. Similarly, direct binding of S1-RBD to recombinant human α v β 3 and α v β 6 integrins, but not α 5 β 1 integrins, was observed by surface plasmon resonance. Adhesion to S1-RBD initiated cell spreading, focal adhesion formation, and actin stress fiber organization to a similar extent as fibronectin. Moreover, S1-RBD stimulated tyrosine phosphorylation of the adhesion mediators FAK, Src, and paxillin, Akt activation, and supported cell proliferation. Together, these data demonstrate that the RGD sequence within S1-RBD can function as an α v -selective integrin agonist. This study provides evidence that cell surface α v -containing integrins can respond functionally to spike protein and raise the possibility that S1-mediated dysregulation of ECM dynamics may contribute to the pathogenesis and/or post-acute sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emma G. Norris
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Xuan Sabrina Pan
- Department of Biomedical Engineering University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Denise C. Hocking
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Biomedical Engineering University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|