1
|
Yu Z, Li Y, Bai L, Zheng Y, Liu X, Zhen Y. The triple combination DBDx alleviates cytokine storm and related lung injury. Int Immunopharmacol 2024; 143:113431. [PMID: 39454409 DOI: 10.1016/j.intimp.2024.113431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Cytokine storm is a life-threatening disorder, and therapeutic treatments are urgently needed. Here, we investigated the anti-cytokine storm efficacy of DBDx, a triple drug combination composed of dipyridamole, ubenimex and dexamethasone. Evaluated by lipopolysaccharide (LPS)-induced cytokine storm murine model, DBDx significantly improved survival rate and prolonged survival time of the model mice. Notably, the efficacy of DBDx was higher than that of dipyridamole, ubenimex and dexamethasone. Determined by ELISA, DBDx significantly reduced the LPS-stimulated serum levels of TNF-α, IL-6 and IL-1β in mice. Luminex assay showed that DBDx suppressed the serum levels of a wide variety of inflammatory cytokines and chemokines, which was more potent than dexamethasone alone. Otherwise, DBDx exerted similar inhibitory effects on cytokine profiles in bronchoalveolar lavage fluid. Histopathological observation showed that DBDx significantly reduced the LPS-induced thickening of alveolar septum, indicating its suppression of capillary congestion, edema and neutrophil infiltration in the lung. Ultra-structure analysis showed that DBDx suppressed the LPS-induced morphological changes of microvilli in type II pneumocytes. In vitro experiment showed that DBDx inhibited IL-6 and TNF-α secretion in THP-1 cells, and downregulated TLR4/NF-κB/HIF-1α signaling pathway. All of these results demonstrate that DBDx, a triple combination of clinical orally-administered drugs, can alleviate cytokine storm and related lung injury. DBDx is beneficial for treating cytokine storm disorders.
Collapse
Affiliation(s)
- Zhuojun Yu
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Bai
- NHC Key Laboratory of Biotechnology of Antibiotics, CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanbo Zheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiujun Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongsu Zhen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Huynh DTN, Nguyen HT, Hsieh CM. Taiwan Chingguan Yihau may improve post-COVID-19 respiratory complications through PI3K/AKT, HIF-1, and TNF signaling pathways revealed by network pharmacology analysis. Mol Divers 2024:10.1007/s11030-024-10993-8. [PMID: 39382736 DOI: 10.1007/s11030-024-10993-8] [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: 06/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024]
Abstract
The emergence of new SARS-CoV-2 variants with a higher contagious capability and faster transmissible speed has imposed an incessant menace on global health and the economy. The SARS-CoV-2 infection might reoccur and last much longer than expected. Thence, there is a high possibility that COVID-19 can cause long-term health problems. This condition needs to be investigated thoroughly, especially the post-COVID-19 complications. Respiratory tract disorders are common and typical complications after recovery. Until now, there has been a lack of data on specialized therapeutic medicine for post-COVID-19 complications. The clinical efficacy of NRICM101 has been demonstrated in hospitalized COVID-19 patients. This herbal medicine may also be a promising therapy for post-COVID-19 complications, thanks to its phytochemical constituents. The potential pharmacological mechanisms of NRICM101 in treating post-COVID-19 respiratory complications were investigated using network pharmacology combined with molecular docking, and the results revealed that NRICM101 may exert a beneficial effect through the three primary pathways: PI3K/AKT, HIF-1, and TNF signaling pathways. Flavonoids (especially quercetin) have a predominant role and synergize with other active compounds to produce therapeutic effectiveness. Most of the main active compounds exist in three chief herbal ingredients, including Liquorice root (Glycyrrhiza glabra), Scutellaria root (Scutellaria baicalensis), and Mulberry leaf (Morus alba). To our knowledge, this is the first study of the NRICM101 effect on post-COVID-19 respiratory complications. Our findings may provide a better understanding of the potential mechanisms of NRICM101 in treating SARS-CoV-2 infection and regulating the immunoinflammatory response to improve post-COVID-19 respiratory complications.
Collapse
Affiliation(s)
- Dung Tam Nguyen Huynh
- College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Can Tho University of Medicine and Pharmacy, Can Tho City, 94117, Vietnam
| | - Hien Thi Nguyen
- Department of Nutrition and Food Safety, Faculty of Public Health, Can Tho University of Medicine and Pharmacy, Can Tho City, 94117, Vietnam
| | - Chien-Ming Hsieh
- College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
3
|
Pedreañez A, Mosquera-Sulbaran JA, Tene D. Role of the receptor for advanced glycation end products in the severity of SARS-CoV-2 infection in diabetic patients. Diabetol Int 2024; 15:732-744. [PMID: 39469543 PMCID: PMC11512988 DOI: 10.1007/s13340-024-00746-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/17/2024] [Indexed: 10/30/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, is a severe disease in older adults and in individuals with associated comorbidities such as diabetes mellitus. Patients with diabetes infected with SARS-CoV-2 are more likely to develop severe pneumonia, hospitalization, and mortality compared with infected non-diabetic patients. During diabetes, hyperglycemia contributes to the maintenance of a low-grade inflammatory state which has been implicated in the microvascular and macrovascular complications associated with this pathology. The receptor for advanced glycation end products (RAGE) is a multi-ligand pattern recognition receptor, expressed on a wide variety of cells, which participates as an important mediator of inflammatory responses in many diseases, including lung diseases. This review highlights the role of RAGE in the pathophysiology of COVID-19 with special emphasis on diabetic patients. These data could explain the severity of the disease, positioning it as a key therapeutic target in the clinical management of this infection.
Collapse
Affiliation(s)
- Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Apartado Postal: 23, Maracaibo 4001-A, Maracaibo, Zulia Venezuela
| | - Jesús A. Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas “Dr. Américo Negrette”, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Diego Tene
- Universidad Nacional del Chimborazo, Facultad de Ciencias de la Salud, Riobamba, Ecuador
| |
Collapse
|
4
|
Wang D, Wang M, Sun S, Zhang C, Song Y, Li J, Song B, Lv H, Wang S, Jiang W. Hypoxia-induced NLRP3 inflammasome activation via the HIF-1α/NF-κB signaling pathway in human dental pulp fibroblasts. BMC Oral Health 2024; 24:1156. [PMID: 39343901 PMCID: PMC11441079 DOI: 10.1186/s12903-024-04936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Previous studies have reported the link between hypoxic conditions and NLRP3 inflammasome-mediated pulpal inflammation in the progression of pulpitis. However, the underlying mechanism has not been fully elucidated. This study aimed to investigate the role of HIF-1α in the regulation of NLRP3 inflammasome pathway via NF-κB signaling under hypoxic conditions with or without LPS in human dental pulp fibroblasts (HDPFs) during the progression of pulpitis. METHODS HIF-1α plasmids or siRNAs were used to upregulate or downregulate HIF-1α in HDPFs, respectively. The effect of hypoxia with or without LPS on the NF-κB signaling and NLRP3 inflammasome pathway was analyzed by immunofluorescence staining, qRT-PCR, western blotting and ELISA. RESULTS The hypoxic conditions alone induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling in a time-dependent manner in HDPFs. The upregulation of HIF-1α further promoted hypoxia-induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. In comparison, downregulation of HIF-1α inhibited ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. Additionally, LPS plus hypoxia further promoted HIF-1α expression and NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. CONCLUSIONS HIF-1α served as a positive regulator of NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling in HDPFs in the sterile pulpal inflammation and caries-related pulpitis microenvironment. The finding of a novel functional HIF-1α-NF-κB-NLRP3 axis provides insight into the link between the hypoxic microenvironment and pulpal inflammation, thus supporting a promising therapeutic strategy for the control of pulpal inflammation.
Collapse
Affiliation(s)
- Diya Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Minghao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Shukai Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chongyang Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Ya Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jianing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Bing Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Haipeng Lv
- Department of Stomatology, Xi'an Daxing Hospital, Xi'an, Shaanxi, China.
| | - Shengchao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Wenkai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| |
Collapse
|
5
|
Zhao X, Yang L. Pharmacological targets and validation of remdesivir for the treatment of COVID-19-associated pulmonary fibrosis: A network-based pharmacology and bioinformatics study. Medicine (Baltimore) 2024; 103:e39062. [PMID: 39331891 PMCID: PMC11441881 DOI: 10.1097/md.0000000000039062] [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: 09/29/2024] Open
Abstract
The objective of this study was to employ bioinformatics and network pharmacology methodologies to investigate the targets and molecular mechanisms of remdesivir in the treatment of coronavirus disease 2019 (COVID-19)-associated pulmonary fibrosis (PF). Several open-source databases were utilized to confirm the shared targets of remdesivir, COVID-19, and PF. Following this, a comprehensive analysis incorporating function enrichment, protein-protein interaction (PPI), transcription factor (TF), and molecular docking was conducted to investigate the potential mechanisms underlying the effectiveness of remdesivir in the treatment of COVID-19-associated PF. The initial validation of these findings was performed using publicly available histological and single-cell sequencing databases. The functional enrichment analysis revealed a strong association between remdesivir and viral defense, inflammatory response, and immune response. The key pathways identified in the study were transforming growth factor (TGF-β), PI3K-Akt, mTOR, MAPK, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor resistance, HIF-1, and Toll-like receptor signaling pathways. Additionally, the PPI analysis demonstrated the network relationships of 13 important targets, while the TF analysis provided valuable insights into the regulatory networks of these targets. Among the identified TFs, RELA was found to be the most significant. To validate our findings, we utilized publicly available histological and single-cell sequencing databases, successfully confirming the involvement of 8 key targets, including AKT1, EGFR, RHOA, MAPK1, PIK3R1, MAPK8, MAPK14, and MTOR. Furthermore, molecular docking studies were conducted to assess the interaction between remdesivir and the identified key targets, thus confirming its effective targeting effects. Remdesivir has the potential to exert antiviral, anti-inflammatory, and immunomodulatory effects in the context of COVID-19-associated PF.
Collapse
Affiliation(s)
- Xueping Zhao
- Department of Pharmacy, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, China
| | - Liping Yang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing, China
| |
Collapse
|
6
|
Li S, Guo S, Liu F, Yao Y, Zhu Y, Feng WH. miR-451-targeted PSMB8 promotes PRRSV infection by degrading IRF3. J Virol 2024; 98:e0078424. [PMID: 39194214 PMCID: PMC11407001 DOI: 10.1128/jvi.00784-24] [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: 05/01/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Porcine respiratory and reproductive syndrome (PRRS) is one of the most devastating infectious diseases of pigs, causing reproductive failures in sows and severe respiratory symptoms in piglets and growing pigs. MicroRNAs (miRNAs) are reported to play an essential role in virus-host interactions. In this study, we demonstrated that miR-451 enhanced type I interferon (IFN-I) production through targeting proteasome subunit β8 (PSMB8), therefore restricting PRRS virus (PRRSV) replication. We showed that the expression of PSMB8 was upregulated by PRRSV infection, and knockdown of PSMB8 inhibited PRRSV replication by promoting IFN-I production. Moreover, we demonstrated that PSMB8 interacted with the regulatory domain of IRF3 to mediate K48-linked polyubiquitination and degradation of IRF3. Also, importantly, we showed that PSMB8, as a target gene of miR-451, negatively regulated IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to modulate innate immune responses. IMPORTANCE Porcine respiratory and reproductive syndrome virus (PRRSV), as a huge threat to the swine industry, is a causative agent that urgently needs to be solved. The dissecting of PRRSV pathogenesis and understanding of the host-pathogen interaction will provide insights into developing effective anti-PRRSV strategies. In this study, we showed that miR-451 dramatically inhibited PRRSV replication by targeting proteasome subunit β8 (PSMB8), a subunit of the immunoproteasome. Mutation of PSMB8 is often related to autoinflammatory diseases due to the elevated IFN production. We revealed that PSMB8 downregulated IFN production by promoting IRF3 degradation. In addition, we showed that PRRSV infection upregulated PSMB8 expression. Taken together, our findings reveal that miR-451 is a negative regulator of PRRSV replication, and PSMB8, a target gene of miR-451, negatively regulates IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to regulate innate immune responses.
Collapse
Affiliation(s)
- Sihan Li
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shuyuan Guo
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Fang Liu
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yao Yao
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingqi Zhu
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wen-Hai Feng
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Eisenreich W, Leberfing J, Rudel T, Heesemann J, Goebel W. Interactions of SARS-CoV-2 with Human Target Cells-A Metabolic View. Int J Mol Sci 2024; 25:9977. [PMID: 39337465 PMCID: PMC11432161 DOI: 10.3390/ijms25189977] [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: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Viruses are obligate intracellular parasites, and they exploit the cellular pathways and resources of their respective host cells to survive and successfully multiply. The strategies of viruses concerning how to take advantage of the metabolic capabilities of host cells for their own replication can vary considerably. The most common metabolic alterations triggered by viruses affect the central carbon metabolism of infected host cells, in particular glycolysis, the pentose phosphate pathway, and the tricarboxylic acid cycle. The upregulation of these processes is aimed to increase the supply of nucleotides, amino acids, and lipids since these metabolic products are crucial for efficient viral proliferation. In detail, however, this manipulation may affect multiple sites and regulatory mechanisms of host-cell metabolism, depending not only on the specific viruses but also on the type of infected host cells. In this review, we report metabolic situations and reprogramming in different human host cells, tissues, and organs that are favorable for acute and persistent SARS-CoV-2 infection. This knowledge may be fundamental for the development of host-directed therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Julian Leberfing
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Thomas Rudel
- Chair of Microbiology, Biocenter, University of Würzburg, 97074 Würzburg, Germany;
| | - Jürgen Heesemann
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| | - Werner Goebel
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| |
Collapse
|
8
|
Mou X, Luo F, Zhang W, Cheng Q, Hepojoki J, Zhu S, Liu Y, Xiong H, Guo D, Yu J, Chen L, Li Y, Hou W, Chen S. SARS-CoV-2 NSP16 promotes IL-6 production by regulating the stabilization of HIF-1α. Cell Signal 2024; 124:111387. [PMID: 39251053 DOI: 10.1016/j.cellsig.2024.111387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent of coronavirus disease 2019 (COVID-19). Severe and fatal COVID-19 cases often display cytokine storm i.e. significant elevation of pro-inflammatory cytokines and acute respiratory distress syndrome (ARDS) with systemic hypoxia. Understanding the mechanisms of these pathogenic manifestations would be essential for the prevention and especially treatment of COVID-19 patients. Here, using a dual luciferase reporter assay for hypoxia-response element (HRE), we initially identified SARS-CoV-2 nonstructural protein 5 (NSP5), NSP16, and open reading frame 3a (ORF3a) to upregulate hypoxia-inducible factor-1α (HIF-1α) signaling. Further experiments showed NSP16 to have the most prominent effect on HIF-1α, thus contributing to the induction of COVID-19 associated pro-inflammatory response. We demonstrate that NSP16 interrupts von Hippel-Lindau (VHL) protein interaction with HIF-1α, thereby inhibiting ubiquitin-dependent degradation of HIF-1α and allowing it to bind HRE region in the IL-6 promoter region. Taken together, the findings imply that SARS-CoV-2 NSP16 induces HIF-1α expression, which in turn exacerbates the production of IL-6.
Collapse
Affiliation(s)
- Xiaoli Mou
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Fan Luo
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, 00290 Helsinki, Finland
| | - Weihao Zhang
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Qi Cheng
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, 00290 Helsinki, Finland
| | - Shaowei Zhu
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Yuanyuan Liu
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Hairong Xiong
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Deyin Guo
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Jingyou Yu
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Liangjun Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Wei Hou
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; School of Public Health, Wuhan University, Wuhan, Hubei 430071, China; School of Ecology and Environment, Tibet University, Lhasa, Tibet 850000, China; Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong 518057, China.
| | - Shuliang Chen
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China.
| |
Collapse
|
9
|
Ha DP, Shin WJ, Liu Z, Doche ME, Lau R, Leli NM, Conn CS, Russo M, Lorenzato A, Koumenis C, Yu M, Mumenthaler SM, Lee AS. Targeting stress induction of GRP78 by cardiac glycoside oleandrin dually suppresses cancer and COVID-19. Cell Biosci 2024; 14:115. [PMID: 39238058 PMCID: PMC11378597 DOI: 10.1186/s13578-024-01297-3] [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: 04/16/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Despite recent therapeutic advances, combating cancer resistance remains a formidable challenge. The 78-kilodalton glucose-regulated protein (GRP78), a key stress-inducible endoplasmic reticulum (ER) chaperone, plays a crucial role in both cancer cell survival and stress adaptation. GRP78 is also upregulated during SARS-CoV-2 infection and acts as a critical host factor. Recently, we discovered cardiac glycosides (CGs) as novel suppressors of GRP78 stress induction through a high-throughput screen of clinically relevant compound libraries. This study aims to test the possibility that agents capable of blocking stress induction of GRP78 could dually suppress cancer and COVID-19. RESULTS Here we report that oleandrin (OLN), is the most potent among the CGs in inhibiting acute stress induction of total GRP78, which also results in reduced cell surface and nuclear forms of GRP78 in stressed cells. The inhibition of stress induction of GRP78 is at the post-transcriptional level, independent of protein degradation and autophagy and may involve translational control as OLN blocks stress-induced loading of ribosomes onto GRP78 mRNAs. Moreover, the human Na+/K+-ATPase α3 isoform is critical for OLN suppression of GRP78 stress induction. OLN, in nanomolar range, enhances apoptosis, sensitizes colorectal cancer cells to chemotherapeutic agents, and reduces the viability of patient-derived colon cancer organoids. Likewise, OLN, suppresses GRP78 expression and impedes tumor growth in an orthotopic breast cancer xenograft model. Furthermore, OLN blocks infection by SARS-CoV-2 and its variants and enhances existing anti-viral therapies. Notably, GRP78 overexpression mitigates OLN-mediated cancer cell apoptotic onset and suppression of virus release. CONCLUSION Our findings validate GRP78 as a target of OLN anti-cancer and anti-viral activities. These proof-of-principle studies support further investigation of OLN as a readily accessible compound to dually combat cancer and COVID-19.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Woo-Jin Shin
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, 34987, USA
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Michael E Doche
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Roy Lau
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariangela Russo
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Annalisa Lorenzato
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Min Yu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shannon M Mumenthaler
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
10
|
Gao C, Xiao C, Wang M, Liang X, Qin C, Zhang H, Bai R, Zhang R, Feng W, Yang J, Tang J. HIF-1 Transcriptionally Regulates Basal Expression of STING to Maintain Cellular Innate Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:494-505. [PMID: 38967520 DOI: 10.4049/jimmunol.2400123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024]
Abstract
Stimulator of IFN genes (STING) is a critical component of the innate immune system, playing an essential role in defending against DNA virus infections. However, the mechanisms governing basal STING regulation remain poorly understood. In this study, we demonstrate that the basal level of STING is critically maintained by hypoxia-inducible factor 1 (HIF-1)α through transcription. Under normal conditions, HIF-1α binds constitutively to the promoter region of STING, actively promoting its transcription. Knocking down HIF-1α results in a decrease in STING expression in multiple cell lines and zebrafish, which in turn reduces cellular responses to synthetic dsDNAs, including cell signaling and IFN production. Moreover, this decrease in STING levels leads to an increase in cellular susceptibility to DNA viruses HSV-1 and pseudorabies virus. These findings unveil a (to our knowledge) novel role of HIF-1α in maintaining basal STING levels and provide valuable insights into STING-mediated antiviral activities and associated diseases.
Collapse
Affiliation(s)
- Chao Gao
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chenglu Xiao
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengdong Wang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xinxin Liang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chao Qin
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hang Zhang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Rulan Bai
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Rui Zhang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenhai Feng
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jinbo Yang
- Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jun Tang
- National Key Laboratory of Veterinary Public Health Security, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Lin J, Bai S, He L, Yang Y, Li X, Luo L, Wang Y, Chen YY, Qin J, Zhong Y. Cytotoxic Lymphocyte-Monocyte Complex Reflects the Dynamics of Coronavirus Disease 2019 Systemic Immune Response. J Infect Dis 2024; 230:5-14. [PMID: 39052699 DOI: 10.1093/infdis/jiae048] [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: 07/16/2023] [Revised: 12/01/2023] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes a variety of clinical manifestations, many of which originate from altered immune responses, either locally or systemically. Immune cell cross-talk occurs mainly in lymphoid organs. However, systemic cell interaction specific to coronavirus disease 2019 has not been well characterized. Here, by employing single-cell RNA sequencing and imaging flow cytometry analysis, we unraveled, in peripheral blood, a heterogeneous group of cell complexes formed by the adherence of CD14+ monocytes to different cytotoxic lymphocytes, including SARS-CoV-2-specific CD8+ T cells, γδ T cells, and natural killer T cells. These lymphocytes attached to CD14+ monocytes that showed enhanced inflammasome activation and pyroptosis-induced cell death in progression stage; in contrast, in the convalescent phase, CD14+ monocytes with elevated antigen presentation potential were targeted by cytotoxic lymphocytes, thereby restricting the excessive immune activation. Collectively, our study reports previously unrecognized cell-cell interplay in the SARS-CoV-2-specific immune response, providing new insight into the intricacy of dynamic immune cell interaction representing antiviral defense.
Collapse
Affiliation(s)
- Jiajia Lin
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Shiyu Bai
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Liheng He
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
| | - Ye Yang
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Xiyue Li
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Liulin Luo
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine
| | - Ying Wang
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying-Ying Chen
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhong Qin
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
| | - Yi Zhong
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| |
Collapse
|
12
|
Guarnieri JW, Lie T, Albrecht YES, Hewin P, Jurado KA, Widjaja GA, Zhu Y, McManus MJ, Kilbaugh TJ, Keith K, Potluri P, Taylor D, Angelin A, Murdock DG, Wallace DC. Mitochondrial antioxidants abate SARS-COV-2 pathology in mice. Proc Natl Acad Sci U S A 2024; 121:e2321972121. [PMID: 39008677 PMCID: PMC11287122 DOI: 10.1073/pnas.2321972121] [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: 12/13/2023] [Accepted: 06/07/2024] [Indexed: 07/17/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection inhibits mitochondrial oxidative phosphorylation (OXPHOS) and elevates mitochondrial reactive oxygen species (ROS, mROS) which activates hypoxia-inducible factor-1alpha (HIF-1α), shifting metabolism toward glycolysis to drive viral biogenesis but also causing the release of mitochondrial DNA (mtDNA) and activation of innate immunity. To determine whether mitochondrially targeted antioxidants could mitigate these viral effects, we challenged mice expressing human angiotensin-converting enzyme 2 (ACE2) with SARS-CoV-2 and intervened using transgenic and pharmacological mitochondrially targeted catalytic antioxidants. Transgenic expression of mitochondrially targeted catalase (mCAT) or systemic treatment with EUK8 decreased weight loss, clinical severity, and circulating levels of mtDNA; as well as reduced lung levels of HIF-1α, viral proteins, and inflammatory cytokines. RNA-sequencing of infected lungs revealed that mCAT and Eukarion 8 (EUK8) up-regulated OXPHOS gene expression and down-regulated HIF-1α and its target genes as well as innate immune gene expression. These data demonstrate that SARS-CoV-2 pathology can be mitigated by catalytically reducing mROS, potentially providing a unique host-directed pharmacological therapy for COVID-19 which is not subject to viral mutational resistance.
Collapse
Affiliation(s)
- Joseph W. Guarnieri
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Timothy Lie
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- University of Pennsylvania, Philadelphia, PA19104
| | - Yentli E. Soto Albrecht
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Peter Hewin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Kellie A. Jurado
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Gabrielle A. Widjaja
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Yi Zhu
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Meagan J. McManus
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Todd J. Kilbaugh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Kelsey Keith
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Prasanth Potluri
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Deanne Taylor
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Alessia Angelin
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Deborah G. Murdock
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Douglas C. Wallace
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
13
|
Yang S, Tian M, Dai Y, Wang R, Yamada S, Feng S, Wang Y, Chhangani D, Ou T, Li W, Guo X, McAdow J, Rincon-Limas DE, Yin X, Tai W, Cheng G, Johnson A. Infection and chronic disease activate a systemic brain-muscle signaling axis. Sci Immunol 2024; 9:eadm7908. [PMID: 38996009 DOI: 10.1126/sciimmunol.adm7908] [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: 11/06/2023] [Revised: 04/18/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
Infections and neurodegenerative diseases induce neuroinflammation, but affected individuals often show nonneural symptoms including muscle pain and muscle fatigue. The molecular pathways by which neuroinflammation causes pathologies outside the central nervous system (CNS) are poorly understood. We developed multiple models to investigate the impact of CNS stressors on motor function and found that Escherichia coli infections and SARS-CoV-2 protein expression caused reactive oxygen species (ROS) to accumulate in the brain. ROS induced expression of the cytokine Unpaired 3 (Upd3) in Drosophila and its ortholog, IL-6, in mice. CNS-derived Upd3/IL-6 activated the JAK-STAT pathway in skeletal muscle, which caused muscle mitochondrial dysfunction and impaired motor function. We observed similar phenotypes after expressing toxic amyloid-β (Aβ42) in the CNS. Infection and chronic disease therefore activate a systemic brain-muscle signaling axis in which CNS-derived cytokines bypass the connectome and directly regulate muscle physiology, highlighting IL-6 as a therapeutic target to treat disease-associated muscle dysfunction.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Meijie Tian
- Genetics Branch, Oncogenomics Section, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yulong Dai
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Rong Wang
- Department of Genetics and Genetics Engineering, School of Life Science, Fudan University, Shanghai 200438, China
| | - Shigehiro Yamada
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yunyun Wang
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Deepak Chhangani
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Tiffany Ou
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Wenle Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuan Guo
- Life Science Institute, Jinzhou Medical University, Jinzhou 121001, China
| | - Jennifer McAdow
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Diego E Rincon-Limas
- Department of Neurology and McKnight Brain Institute, Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, Genetics Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
- Southwest United Graduate School, Kunming 650092, China
| | - Aaron Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
14
|
Chen Y, Deng H, Zhou R, Jiang X, Wang H, Xin S, Mo W, Wang S, Liu Y. Comprehensive mapping of immune perturbations associated with secondary hemophagocytic lymphohistiocytosis. J Leukoc Biol 2024:qiae138. [PMID: 38973235 DOI: 10.1093/jleuko/qiae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
Secondary hemophagocytic lymphohistiocytosis (sHLH) is a hyperinflammatory syndrome characterized by immune disorders. It is imperative to elucidate the immunophenotypic panorama and the interactions among these cells in patients. Human peripheral blood mononuclear cells were collected from healthy donors and sHLH patients and tested using multicolor flow cytometry. We used FlowSOM to explore and visualize the immunophenotypic characteristics of sHLH. By demonstrating the phenotypes of immune cells, we discovered that sHLH patients had significantly higher levels of CD56+ monocytes, higher levels of myeloid-derived suppressor cells, low-density neutrophil-to-T cell ratio, and higher heterogeneous T cell activation than healthy donors. However, natural killer cell cytotoxicity and function were impaired. We then assessed the correlations among 30 immune cell types and evaluated metabolic analysis. Our findings demonstrated polymorphonuclear myeloid-derived suppressor cells, CD56+ monocytes, and neutrophil-to-T cell ratio were elevated abnormally in sHLH patients, which may indicate an association with immune overactivation and inflammatory response. We are expected to confirm that they are involved in the occurrence of the disease through further in-depth research.
Collapse
Affiliation(s)
- Yinchun Chen
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Haimei Deng
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628, Zhenyuan Road, Xinhuling Street, Shenzhen 518118, China
| | - Ruiqing Zhou
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Xiaotao Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Guangzhou 510400, China
| | - Huijuan Wang
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, No. 10 Huan Yu Second Road, Guangzhou 510180, China
| | - Songqing Xin
- Changan Hospital of Dongguan, No. 171 Changqing South Road, Dongguan 523850, China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Shunqing Wang
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Yufeng Liu
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, No. 10 Huan Yu Second Road, Guangzhou 510180, China
| |
Collapse
|
15
|
Shouman S, El-Kholy N, Hussien AE, El-Derby AM, Magdy S, Abou-Shanab AM, Elmehrath AO, Abdelwaly A, Helal M, El-Badri N. SARS-CoV-2-associated lymphopenia: possible mechanisms and the role of CD147. Cell Commun Signal 2024; 22:349. [PMID: 38965547 PMCID: PMC11223399 DOI: 10.1186/s12964-024-01718-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.
Collapse
Affiliation(s)
- Shaimaa Shouman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Nada El-Kholy
- Department of Drug Discovery, H. Lee Moffit Cancer Center& Research Institute, Tampa, FL, 33612, USA
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Alaa E Hussien
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | | | - Ahmad Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Mohamed Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt.
| |
Collapse
|
16
|
Chang CC, Wu CY, Wu CM, Wu CW, Wang YC, Lin GJ, Chien MS, Huang C. Cytotoxicity effect and transcriptome analysis of PCV3-infected cells revealed potential viral pathogenic mechanisms. Microb Pathog 2024; 192:106715. [PMID: 38810767 DOI: 10.1016/j.micpath.2024.106715] [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: 01/24/2024] [Revised: 05/10/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Porcine circovirus type 3 (PCV3) has become an important pathogen in the global swine industry and poses a threat to pig health, but its pathogenic mechanism remains unknown. In this study, we constructed an innovative, linear infectious clone of PCV3 for rescuing the virus, and explored the transcriptome of infected cells to gain insights into its pathogenic mechanisms. Subsequently, an in vivo experiment was conducted to evaluate the pathogenicity of the rescued virus in pig. PCV3 nucleic acid was distributed across various organs, indicating systemic circulation via the bloodstream and viremia. Immunohistochemical staining also revealed a significant presence of PCV3 antigens in the spleen, lungs, and lymph nodes, indicating that PCV3 had tropism for these organs. Transcriptome analysis of infected ST cells revealed differential expression of genes associated with apoptosis, immune responses, and cellular metabolism. Notably, upregulation of genes related to the hypoxia-inducible factor-1 pathway, glycolysis, and the AGE/RAGE pathway suggests activation of inflammatory responses, ultimately leading to onset of disease. These findings have expanded our understanding of PCV3 pathogenesis, and the interplay between PCV3 and host factors.
Collapse
Affiliation(s)
- Chia-Chun Chang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Ching-Ying Wu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Chi-Ming Wu
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Ching-Wei Wu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan; Research Center for Animal Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Yi-Chen Wang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Guang-Jan Lin
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Maw-Sheng Chien
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan; Research Center for Animal Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan.
| | - Chienjin Huang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan.
| |
Collapse
|
17
|
Han H, Kim JE, Lee HJ. Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model. Sci Rep 2024; 14:14545. [PMID: 38914619 PMCID: PMC11196261 DOI: 10.1038/s41598-024-65447-w] [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: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.
Collapse
Affiliation(s)
- Hengmin Han
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Jung-Eun Kim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
18
|
Rurek M. Mitochondria in COVID-19: from cellular and molecular perspective. Front Physiol 2024; 15:1406635. [PMID: 38974521 PMCID: PMC11224649 DOI: 10.3389/fphys.2024.1406635] [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: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
The rapid development of the COVID-19 pandemic resulted in a closer analysis of cell functioning during β-coronavirus infection. This review will describe evidence for COVID-19 as a syndrome with a strong, albeit still underestimated, mitochondrial component. Due to the sensitivity of host mitochondria to coronavirus infection, SARS-CoV-2 affects mitochondrial signaling, modulates the immune response, modifies cellular energy metabolism, induces apoptosis and ageing, worsening COVID-19 symptoms which can sometimes be fatal. Various aberrations across human systems and tissues and their relationships with mitochondria were reported. In this review, particular attention is given to characterization of multiple alterations in gene expression pattern and mitochondrial metabolism in COVID-19; the complexity of interactions between SARS-CoV-2 and mitochondrial proteins is presented. The participation of mitogenome fragments in cell signaling and the occurrence of SARS-CoV-2 subgenomic RNA within membranous compartments, including mitochondria is widely discussed. As SARS-CoV-2 severely affects the quality system of mitochondria, the cellular background for aberrations in mitochondrial dynamics in COVID-19 is additionally characterized. Finally, perspectives on the mitigation of COVID-19 symptoms by affecting mitochondrial biogenesis by numerous compounds and therapeutic treatments are briefly outlined.
Collapse
Affiliation(s)
- Michał Rurek
- Department of Molecular and Cellular Biology, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
19
|
Zhang Y, Yang N, Li Y, Tan C, Cai Y, Rui X, Liu Y, Fu Y, Liu G. Transmissible gastroenteritis virus induces inflammatory responses via RIG-I/NF-κB/HIF-1α/glycolysis axis in intestinal organoids and in vivo. J Virol 2024; 98:e0046124. [PMID: 38780247 PMCID: PMC11237398 DOI: 10.1128/jvi.00461-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Transmissible gastroenteritis virus (TGEV)-induced enteritis is characterized by watery diarrhea, vomiting, and dehydration, and has high mortality in newborn piglets, resulting in significant economic losses in the pig industry worldwide. Conventional cell lines have been used for many years to investigate inflammation induced by TGEV, but these cell lines may not mimic the actual intestinal environment, making it difficult to obtain accurate results. In this study, apical-out porcine intestinal organoids were employed to study TEGV-induced inflammation. We found that apical-out organoids were susceptible to TGEV infection, and the expression of representative inflammatory cytokines was significantly upregulated upon TGEV infection. In addition, retinoic acid-inducible gene I (RIG-I) and the nuclear factor-kappa B (NF-κB) pathway were responsible for the expression of inflammatory cytokines induced by TGEV infection. We also discovered that the transcription factor hypoxia-inducible factor-1α (HIF-1α) positively regulated TGEV-induced inflammation by activating glycolysis in apical-out organoids, and pig experiments identified the same molecular mechanism as the ex vivo results. Collectively, we unveiled that the inflammatory responses induced by TGEV were modulated via the RIG-I/NF-κB/HIF-1α/glycolysis axis ex vivo and in vivo. This study provides novel insights into TGEV-induced enteritis and verifies intestinal organoids as a reliable model for investigating virus-induced inflammation. IMPORTANCE Intestinal organoids are a newly developed culture system for investigating immune responses to virus infection. This culture model better represents the physiological environment compared with well-established cell lines. In this study, we discovered that inflammatory responses induced by TGEV infection were regulated by the RIG-I/NF-κB/HIF-1α/glycolysis axis in apical-out porcine organoids and in pigs. Our findings contribute to understanding the mechanism of intestinal inflammation upon viral infection and highlight apical-out organoids as a physiological model to mimic virus-induced inflammation.
Collapse
Affiliation(s)
- Yunhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Ning Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Chen Tan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yifei Cai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- Nutritional Biology, Wageningen University and Research, Wageningen, the Netherlands
| | - Xue Rui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yuanyuan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| |
Collapse
|
20
|
Wicik Z, Eyileten C, Nowak A, Keshwani D, Simões SN, Martins DC, Klos K, Wlodarczyk W, Assinger A, Soldacki D, Chcialowski A, Siller-Matula JM, Postula M. Alteration of circulating ACE2-network related microRNAs in patients with COVID-19. Sci Rep 2024; 14:13573. [PMID: 38866792 PMCID: PMC11169442 DOI: 10.1038/s41598-024-58037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/25/2024] [Indexed: 06/14/2024] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) serves as the primary receptor for the SARS-CoV-2 virus and has implications for the functioning of the cardiovascular system. Based on our previously published bioinformatic analysis, in this study we aimed to analyze the diagnostic and predictive utility of miRNAs (miR-10b-5p, miR-124-3p, miR-200b-3p, miR-26b-5p, miR-302c-5p) identified as top regulators of ACE2 network with potential to affect cardiomyocytes and cardiovascular system in patients with COVID-19. The expression of miRNAs was determined through qRT-PCR in a cohort of 79 hospitalized COVID-19 patients as well as 32 healthy volunteers. Blood samples and clinical data of COVID-19 patients were collected at admission, 7-days and 21-days after admission. We also performed SHAP analysis of clinical data and miRNAs target predictions and advanced enrichment analyses. Low expression of miR-200b-3p at the seventh day of admission is indicative of predictive value in determining the length of hospital stay and/or the likelihood of mortality, as shown in ROC curve analysis with an AUC of 0.730 and a p-value of 0.002. MiR-26b-5p expression levels in COVID-19 patients were lower at the baseline, 7 and 21-days of admission compared to the healthy controls (P < 0.0001). Similarly, miR-10b-5p expression levels were lower at the baseline and 21-days post admission (P = 0.001). The opposite situation was observed in miR-124-3p and miR-302c-5p. Enrichment analysis showed influence of analyzed miRNAs on IL-2 signaling pathway and multiple cardiovascular diseases through COVID-19-related targets. Moreover, the COVID-19-related genes regulated by miR-200b-3p were linked to T cell protein tyrosine phosphatase and the HIF-1 transcriptional activity in hypoxia. Analysis focused on COVID-19 associated genes showed that all analyzed miRNAs are strongly affecting disease pathways related to CVDs which could be explained by their strong interaction with the ACE2 network.
Collapse
Affiliation(s)
- Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Center for Preclinical Research and Technology CEPT, Medical University of Warsaw, 02-097, Warsaw, Poland
- Department of Neurochemistry, Institute of Psychiatry and Neurology, Sobieskiego 9 Street, 02-957, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Center for Preclinical Research and Technology CEPT, Medical University of Warsaw, 02-097, Warsaw, Poland
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Anna Nowak
- Department of Experimental and Clinical Pharmacology, Center for Preclinical Research and Technology CEPT, Medical University of Warsaw, 02-097, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091, Warsaw, Poland
- Department of Diabetology and Internal Medicine, University Clinical Centre, Medical University of Warsaw, Warsaw, Poland
| | - Disha Keshwani
- Department of Experimental and Clinical Pharmacology, Center for Preclinical Research and Technology CEPT, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Sérgio N Simões
- Federal Institute of Education, Science and Technology of Espírito Santo, Serra, Espírito Santo, 29056-264, Brazil
| | - David C Martins
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo Andre, 09606-045, Brazil
| | - Krzysztof Klos
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Wojciech Wlodarczyk
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Dariusz Soldacki
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Chcialowski
- Department of Infectious Diseases and Allergology - Military Institute of Medicine, Warsaw, Poland
| | - Jolanta M Siller-Matula
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090, Vienna, Austria
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Center for Preclinical Research and Technology CEPT, Medical University of Warsaw, 02-097, Warsaw, Poland.
| |
Collapse
|
21
|
Guarnieri JW, Haltom JA, Albrecht YES, Lie T, Olali AZ, Widjaja GA, Ranshing SS, Angelin A, Murdock D, Wallace DC. SARS-CoV-2 mitochondrial metabolic and epigenomic reprogramming in COVID-19. Pharmacol Res 2024; 204:107170. [PMID: 38614374 DOI: 10.1016/j.phrs.2024.107170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
To determine the effects of SARS-CoV-2 infection on cellular metabolism, we conducted an exhaustive survey of the cellular metabolic pathways modulated by SARS-CoV-2 infection and confirmed their importance for SARS-CoV-2 propagation by cataloging the effects of specific pathway inhibitors. This revealed that SARS-CoV-2 strongly inhibits mitochondrial oxidative phosphorylation (OXPHOS) resulting in increased mitochondrial reactive oxygen species (mROS) production. The elevated mROS stabilizes HIF-1α which redirects carbon molecules from mitochondrial oxidation through glycolysis and the pentose phosphate pathway (PPP) to provide substrates for viral biogenesis. mROS also induces the release of mitochondrial DNA (mtDNA) which activates innate immunity. The restructuring of cellular energy metabolism is mediated in part by SARS-CoV-2 Orf8 and Orf10 whose expression restructures nuclear DNA (nDNA) and mtDNA OXPHOS gene expression. These viral proteins likely alter the epigenome, either by directly altering histone modifications or by modulating mitochondrial metabolite substrates of epigenome modification enzymes, potentially silencing OXPHOS gene expression and contributing to long-COVID.
Collapse
Affiliation(s)
- Joseph W Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jeffrey A Haltom
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yentli E Soto Albrecht
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy Lie
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arnold Z Olali
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Gabrielle A Widjaja
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sujata S Ranshing
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alessia Angelin
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Deborah Murdock
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Pilger BI, Castro A, Vasconcellos FF, Moura KF, Signini ÉDF, Marqueze LFB, Fiorenza-Neto EA, Rocha MT, Pedroso GS, Cavaglieri CR, Ferreira AG, Figueiredo C, Minuzzi LG, Gatti da Silva GH, Castro GS, Lira FS, Seelaender M, Pinho RA. Obesity-dependent molecular alterations in fatal COVID-19: A retrospective postmortem study of metabolomic profile of adipose tissue. J Cell Biochem 2024; 125:e30566. [PMID: 38591648 DOI: 10.1002/jcb.30566] [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: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
We investigated the effects of obesity on metabolic, inflammatory, and oxidative stress parameters in the adipose tissue of patients with fatal COVID-19. Postmortem biopsies of subcutaneous adipose tissue were obtained from 25 unvaccinated inpatients who passed from COVID-19, stratified as nonobese (N-OB; body mass index [BMI], 26.5 ± 2.3 kg m-2) or obese (OB BMI 34.2 ± 5.1 kg m-2). Univariate and multivariate analyses revealed that body composition was responsible for most of the variations detected in the metabolome, with greater dispersion observed in the OB group. Fifteen metabolites were major segregation factors. Results from the OB group showed higher levels of creatinine, myo-inositol, O-acetylcholine, and succinate, and lower levels of sarcosine. The N-OB group showed lower levels of glutathione peroxidase activity, as well as higher content of IL-6 and adiponectin. We revealed significant changes in the metabolomic profile of the adipose tissue in fatal COVID-19 cases, with high adiposity playing a key role in these observed variations. These findings highlight the potential involvement of metabolic and inflammatory pathways, possibly dependent on hypoxia, shedding light on the impact of obesity on disease pathogenesis and suggesting avenues for further research and possible therapeutic targets.
Collapse
Affiliation(s)
- Bruna I Pilger
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Alex Castro
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
- Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Franciane F Vasconcellos
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Karen F Moura
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Étore De Favari Signini
- Cardiovascular Physical Therapy Laboratory, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Luis Felipe B Marqueze
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Edson A Fiorenza-Neto
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Mateus T Rocha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Giulia S Pedroso
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Antonio G Ferreira
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Luciele G Minuzzi
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Guilherme H Gatti da Silva
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Gabriela S Castro
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Fábio S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Ricardo A Pinho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
23
|
Dehghan Z, Mirmotalebisohi SA, Mozafar M, Sameni M, Saberi F, Derakhshanfar A, Moaedi J, Zohrevand H, Zali H. Deciphering the similarities and disparities of molecular mechanisms behind respiratory epithelium response to HCoV-229E and SARS-CoV-2 and drug repurposing, a systems biology approach. Daru 2024; 32:215-235. [PMID: 38652363 PMCID: PMC11087451 DOI: 10.1007/s40199-024-00507-0] [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: 09/17/2022] [Accepted: 02/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Identifying the molecular mechanisms behind SARS-CoV-2 disparities and similarities will help find new treatments. The present study determines networks' shared and non-shared (specific) crucial elements in response to HCoV-229E and SARS-CoV-2 viruses to recommend candidate medications. METHODS We retrieved the omics data on respiratory cells infected with HCoV-229E and SARS-CoV-2, constructed PPIN and GRN, and detected clusters and motifs. Using a drug-gene interaction network, we determined the similarities and disparities of mechanisms behind their host response and drug-repurposed. RESULTS CXCL1, KLHL21, SMAD3, HIF1A, and STAT1 were the shared DEGs between both viruses' protein-protein interaction network (PPIN) and gene regulatory network (GRN). The NPM1 was a specific critical node for HCoV-229E and was a Hub-Bottleneck shared between PPI and GRN in HCoV-229E. The HLA-F, ADCY5, TRIM14, RPF1, and FGA were the seed proteins in subnetworks of the SARS-CoV-2 PPI network, and HSPA1A and RPL26 proteins were the seed in subnetworks of the PPI network of HCOV-229E. TRIM14, STAT2, and HLA-F played the same role for SARS-CoV-2. Top enriched KEGG pathways included cell cycle and proteasome in HCoV-229E and RIG-I-like receptor, Chemokine, Cytokine-cytokine, NOD-like receptor, and TNF signaling pathways in SARS-CoV-2. We suggest some candidate medications for COVID-19 patient lungs, including Noscapine, Isoetharine mesylate, Cycloserine, Ethamsylate, Cetylpyridinium, Tretinoin, Ixazomib, Vorinostat, Venetoclax, Vorinostat, Ixazomib, Venetoclax, and epoetin alfa for further in-vitro and in-vivo investigations. CONCLUSION We suggested CXCL1, KLHL21, SMAD3, HIF1A, and STAT1, ADCY5, TRIM14, RPF1, and FGA, STAT2, and HLA-F as critical genes and Cetylpyridinium, Cycloserine, Noscapine, Ethamsylate, Epoetin alfa, Isoetharine mesylate, Ribavirin, and Tretinoin drugs to study further their importance in treating COVID-19 lung complications.
Collapse
Affiliation(s)
- Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mozafar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Sameni
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saberi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Derakhshanfar
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Javad Moaedi
- Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Zohrevand
- Student Research Committee, Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Golden TN, Mani S, Linn RL, Leite R, Trigg NA, Wilson A, Anton L, Mainigi M, Conine CC, Kaufman BA, Strauss JF, Parry S, Simmons RA. Extracellular vesicles alter trophoblast function in pregnancies complicated by COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.17.580824. [PMID: 38464046 PMCID: PMC10925147 DOI: 10.1101/2024.02.17.580824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and resulting coronavirus disease (COVID-19) causes placental dysfunction, which increases the risk of adverse pregnancy outcomes. While abnormal placental pathology resulting from COVID-19 is common, direct infection of the placenta is rare. This suggests that pathophysiology associated with maternal COVID-19, rather than direct placental infection, is responsible for placental dysfunction and alteration of the placental transcriptome. We hypothesized that maternal circulating extracellular vesicles (EVs), altered by COVID-19 during pregnancy, contribute to placental dysfunction. To examine this hypothesis, we characterized maternal circulating EVs from pregnancies complicated by COVID-19 and tested their effects on trophoblast cell physiology in vitro . We found that the gestational timing of COVID-19 is a major determinant of circulating EV function and cargo. In vitro trophoblast exposure to EVs isolated from patients with an active infection at the time of delivery, but not EVs isolated from Controls, altered key trophoblast functions including hormone production and invasion. Thus, circulating EVs from participants with an active infection, both symptomatic and asymptomatic cases, can disrupt vital trophoblast functions. EV cargo differed between participants with COVID-19 and Controls, which may contribute to the disruption of the placental transcriptome and morphology. Our findings show that COVID-19 can have effects throughout pregnancy on circulating EVs and circulating EVs are likely to participate in placental dysfunction induced by COVID-19.
Collapse
|
25
|
You J, Huang R, Zhong R, Shen J, Huang S, Chen J, Chen F, Kang Y, Chen L. Serum AXL is a potential molecular marker for predicting COVID-19 progression. Front Immunol 2024; 15:1394429. [PMID: 38799467 PMCID: PMC11116689 DOI: 10.3389/fimmu.2024.1394429] [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: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Background The severity, symptoms, and outcome of COVID-19 is thought to be closely linked to how the virus enters host cells. This process involves the key roles of angiotensin-converting enzyme 2 (ACE2) and the Tyrosine protein kinase receptor UFO (AXL) receptors. However, there is limited research on the circulating levels of ACE2 and AXL and their implications in COVID-19. Methods A control group of 71 uninfected individuals was also included in the study. According to the Guidance for Corona Virus Disease 2019 (10th edition), a cohort of 358 COVID-19 patients were categorized into non-severe and severe cases. Serum ACE2/AXL levels in COVID-19 patients were detected by enzyme-linked immunosorbent assay (ELISA) at different time points post-COVID-19 infection, including days 0-7, 8-15, 31-179 and >180 days. Serum SARS-CoV-2 IgG/IgM antibodies in COVID-19 patients at the same intervals were assessed by using an iFlash 3000 Chemiluminescence Immunoassay Analyzer. The receiver operating characteristic (ROC) curves were used to assess the diagnostic value of the biological markers, and the association between laboratory parameters and illness progression were explored. Results Compared with the uninfected group, the levels of ACE2 and AXL in the COVID-19 group were decreased, and the SARS-COV-2 IgG level was increased. AXL (AUC = 0.774) demonstrated a stronger predictive ability for COVID-19 than ACE2. In the first week after infection, only the level of AXL was statistically different between severe group and non-severe group. After first week, the levels of ACE2 and AXL were different in two groups. Moreover, in severe COVID-19 cases, the serum ACE2, AXL, and SARS-COV-2 IgM levels reached a peak during days 8-15 before declining, whereas serum SARS-COV-2 IgG levels continued to rise, reaching a peak at day 31-180 days before decreasing. In addition, the AXL level continued to decrease and the SARS-COV-2 IgG level continued to increase in the infected group after 180 days compared to the uninfected group. Conclusions The levels of serum ACE2 and AXL correlate with COVID-19 severity. However, AXL can also provide early warning of clinical deterioration in the first week after infection. AXL appears to be a superior potential molecular marker for predicting COVID-19 progression.
Collapse
Affiliation(s)
- Jianbin You
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Rong Huang
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Ruifang Zhong
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Jing Shen
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Shuhang Huang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Chen
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Falin Chen
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Yanli Kang
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Liangyuan Chen
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
26
|
Das S, Khan R, Banerjee S, Ray S, Ray S. Alterations in Circadian Rhythms, Sleep, and Physical Activity in COVID-19: Mechanisms, Interventions, and Lessons for the Future. Mol Neurobiol 2024:10.1007/s12035-024-04178-5. [PMID: 38702566 DOI: 10.1007/s12035-024-04178-5] [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: 10/20/2023] [Accepted: 04/04/2024] [Indexed: 05/06/2024]
Abstract
Although the world is acquitting from the throes of COVID-19 and returning to the regularity of life, its effects on physical and mental health are prominently evident in the post-pandemic era. The pandemic subjected us to inadequate sleep and physical activities, stress, irregular eating patterns, and work hours beyond the regular rest-activity cycle. Thus, perturbing the synchrony of the regular circadian clock functions led to chronic psychiatric and neurological disorders and poor immunological response in several COVID-19 survivors. Understanding the links between the host immune system and viral replication machinery from a clock-infection biology perspective promises novel avenues of intervention. Behavioral improvements in our daily lifestyle can reduce the severity and expedite the convalescent stage of COVID-19 by maintaining consistent eating, sleep, and physical activity schedules. Including dietary supplements and nutraceuticals with prophylactic value aids in combating COVID-19, as their deficiency can lead to a higher risk of infection, vulnerability, and severity of COVID-19. Thus, besides developing therapeutic measures, perpetual healthy practices could also contribute to combating the upcoming pandemics. This review highlights the impact of the COVID-19 pandemic on biological rhythms, sleep-wake cycles, physical activities, and eating patterns and how those disruptions possibly contribute to the response, severity, and outcome of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sandip Das
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Rajni Khan
- National Institute of Pharmaceutical Education and Research (NIPER) - Hajipur, Vaishali, Hajipur, 844102, Bihar, India
| | - Srishti Banerjee
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India.
| |
Collapse
|
27
|
Lopuhaä BV, Guzel C, van der Lee A, van den Bosch TPP, van Kemenade FJ, Huisman MV, Kruip MJHA, Luider TM, von der Thüsen JH. Increase in venous thromboembolism in SARS-CoV-2 infected lung tissue: proteome analysis of lung parenchyma, isolated endothelium, and thrombi. Histopathology 2024; 84:967-982. [PMID: 38253958 DOI: 10.1111/his.15143] [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: 07/13/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
AIMS COVID-19 pneumonia is characterized by an increased rate of deep venous thrombosis and pulmonary embolism. To better understand the pathophysiology behind thrombosis in COVID-19, we performed proteomics analysis on SARS-CoV-2 infected lung tissue. METHODS Liquid chromatography mass spectrometry was performed on SARS-CoV-2 infected postmortem lung tissue samples. Five protein profiling analyses were performed: whole slide lung parenchyma analysis, followed by analysis of isolated thrombi and endothelium, both stratified by disease (COVID-19 versus influenza) and thrombus morphology (embolism versus in situ). Influenza autopsy cases with pulmonary thrombi were used as controls. RESULTS Compared to influenza controls, both analyses of COVID-19 whole-tissue and isolated endothelium showed upregulation of proteins and pathways related to liver metabolism including urea cycle activation, with arginase being among the top upregulated proteins in COVID-19 lung tissue. Analysis of isolated COVID-19 thrombi showed significant downregulation of pathways related to platelet activation compared to influenza thrombi. Analysis of isolated thrombi based on histomorphology shows that in situ thrombi have significant upregulation of coronavirus pathogenesis proteins. CONCLUSIONS The decrease in platelet activation pathways in severe COVID-19 thrombi suggests a relative increase in venous thromboembolism, as thrombi from venous origin tend to contain fewer platelets than arterial thrombi. Based on histomorphology, in situ thrombi show upregulation of various proteins related to SARS-CoV-2 pathogenesis compared to thromboemboli, which may indicate increased in situ pulmonary thrombosis in COVID-19. Therefore, this study supports the increase of venous thromboembolism without undercutting the involvement of in situ thrombosis in severe COVID-19.
Collapse
Affiliation(s)
- Boaz V Lopuhaä
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Coşkun Guzel
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | | | | | | | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke J H A Kruip
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
28
|
Zhang Y, Rui X, Li Y, Zhang Y, Cai Y, Tan C, Yang N, Liu Y, Fu Y, Liu G. Hypoxia inducible factor-1α facilitates transmissible gastroenteritis virus replication by inhibiting type I and type III interferon production. Vet Microbiol 2024; 292:110055. [PMID: 38513523 DOI: 10.1016/j.vetmic.2024.110055] [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: 10/29/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Transmissible gastroenteritis virus (TGEV) is characterized by watery diarrhea, vomiting, and dehydration and is associated with high mortality especially in newborn piglets, causing significant economic losses to the global pig industry. Hypoxia inducible factor-1α (HIF-1α) has been identified as a key regulator of TGEV-induced inflammation, but understanding of the effect of HIF-1α on TGEV infection remains limited. This study found that TGEV infection was associated with a marked increase in HIF-1α expression in ST cells and an intestinal organoid epithelial monolayer. Furthermore, HIF-1α was shown to facilitate TGEV infection by targeting viral replication, which was achieved by restraining type I and type III interferon (IFN) production. In vivo experiments in piglets demonstrated that the HIF-1α inhibitor BAY87-2243 significantly reduced HIF-1α expression and inhibited TGEV replication and pathogenesis by activating IFN production. In summary, we unveiled that HIF-1α facilitates TGEV replication by restraining type I and type III IFN production in vitro, ex vivo, and in vivo. The findings from this study suggest that HIF-1α could be a novel antiviral target and candidate drug against TGEV infection.
Collapse
Affiliation(s)
- Yunhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Belgium; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China
| | - Xue Rui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China; College of Veterinary Medicine, Xinjiang Agricultural University, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Yue Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Yifei Cai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China; Nutritional Biology, Wageningen University and Research, Wageningen, the Netherlands
| | - Chen Tan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Belgium; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China
| | - Ning Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Belgium; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China
| | - Yuanyuan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China; College of Veterinary Medicine, Xinjiang Agricultural University, China
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, China; College of Veterinary Medicine, Xinjiang Agricultural University, China.
| |
Collapse
|
29
|
He S, Liu SQ, Teng XY, He JY, Liu Y, Gao JH, Wu Y, Hu W, Dong ZJ, Bei JX, Xu JH. Comparative single-cell RNA sequencing analysis of immune response to inactivated vaccine and natural SARS-CoV-2 infection. J Med Virol 2024; 96:e29577. [PMID: 38572977 DOI: 10.1002/jmv.29577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/02/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Uncovering the immune response to an inactivated SARS-CoV-2 vaccine (In-Vac) and natural infection is crucial for comprehending COVID-19 immunology. Here we conducted an integrated analysis of single-cell RNA sequencing (scRNA-seq) data from serial peripheral blood mononuclear cell (PBMC) samples derived from 12 individuals receiving In-Vac compared with those from COVID-19 patients. Our study reveals that In-Vac induces subtle immunological changes in PBMC, including cell proportions and transcriptomes, compared with profound changes for natural infection. In-Vac modestly upregulates IFN-α but downregulates NF-κB pathways, while natural infection triggers hyperactive IFN-α and NF-κB pathways. Both In-Vac and natural infection alter T/B cell receptor repertoires, but COVID-19 has more significant change in preferential VJ gene, indicating a vigorous immune response. Our study reveals distinct patterns of cellular communications, including a selective activation of IL-15RA/IL-15 receptor pathway after In-Vac boost, suggesting its potential role in enhancing In-Vac-induced immunity. Collectively, our study illuminates multifaceted immune responses to In-Vac and natural infection, providing insights for optimizing SARS-CoV-2 vaccine efficacy.
Collapse
Affiliation(s)
- Shuai He
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang-Yun Teng
- Medical Laboratory Center, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming, China
| | - Jin-Yong He
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jia-Hui Gao
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Yue Wu
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Wei Hu
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
| | - Zhong-Jun Dong
- School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-Hua Xu
- Medical Laboratory Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, China
- Medical Laboratory Center, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming, China
| |
Collapse
|
30
|
Luo H, Yan J, Gong R, Zhang D, Zhou X, Wang X. Identification of biomarkers and pathways for the SARS-CoV-2 infections in obstructive sleep apnea patients based on machine learning and proteomic analysis. BMC Pulm Med 2024; 24:112. [PMID: 38443855 PMCID: PMC10913609 DOI: 10.1186/s12890-024-02921-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND The prevalence of obstructive sleep apnea (OSA) was found to be higher in individuals following COVID-19 infection. However, the intricate mechanisms that underscore this concomitance remain partially elucidated. The aim of this study was to delve deeper into the molecular mechanisms that underpin this comorbidity. METHODS We acquired gene expression profiles for COVID-19 (GSE157103) and OSA (GSE75097) from the Gene Expression Omnibus (GEO) database. Upon identifying shared feature genes between OSA and COVID-19 utilizing LASSO, Random forest and Support vector machines algorithms, we advanced to functional annotation, analysis of protein-protein interaction networks, module construction, and identification of pivotal genes. Furthermore, we established regulatory networks encompassing transcription factor (TF)-gene and TF-miRNA interactions, and searched for promising drug targets. Subsequently, the expression levels of pivotal genes were validated through proteomics data from COVID-19 cases. RESULTS Fourteen feature genes shared between OSA and COVID-19 were selected for further investigation. Through functional annotation, it was indicated that metabolic pathways play a role in the pathogenesis of both disorders. Subsequently, employing the cytoHubba plugin, ten hub genes were recognized, namely TP53, CCND1, MDM2, RB1, HIF1A, EP300, STAT3, CDK2, HSP90AA1, and PPARG. The finding of proteomics unveiled a substantial augmentation in the expression level of HSP90AA1 in COVID-19 patient samples, especially in severe conditions. CONCLUSIONS Our investigation illuminate a mutual pathogenic mechanism that underlies both OSA and COVID-19, which may provide novel perspectives for future investigations into the underlying mechanisms.
Collapse
Affiliation(s)
- Hong Luo
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jisong Yan
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Gong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, China
| | - Dingyu Zhang
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, China
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Xia Zhou
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China.
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China.
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China.
| | - Xianguang Wang
- Department of Tuberculosis and Respiratory, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China.
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China.
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
31
|
Lempesis IG, Georgakopoulou VE, Reiter RJ, Spandidos DA. A mid‑pandemic night's dream: Melatonin, from harbinger of anti‑inflammation to mitochondrial savior in acute and long COVID‑19 (Review). Int J Mol Med 2024; 53:28. [PMID: 38299237 PMCID: PMC10852014 DOI: 10.3892/ijmm.2024.5352] [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: 10/11/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
Coronavirus disease 2019 (COVID‑19), a systemic illness caused by severe acute respiratory distress syndrome 2 (SARS‑CoV‑2), has triggered a worldwide pandemic with symptoms ranging from asymptomatic to chronic, affecting practically every organ. Melatonin, an ancient antioxidant found in all living organisms, has been suggested as a safe and effective therapeutic option for the treatment of SARS‑CoV‑2 infection due to its good safety characteristics and broad‑spectrum antiviral medication properties. Melatonin is essential in various metabolic pathways and governs physiological processes, such as the sleep‑wake cycle and circadian rhythms. It exhibits oncostatic, anti‑inflammatory, antioxidant and anti‑aging properties, exhibiting promise for use in the treatment of numerous disorders, including COVID‑19. The preventive and therapeutic effects of melatonin have been widely explored in a number of conditions and have been well‑established in experimental ischemia/reperfusion investigations, particularly in coronary heart disease and stroke. Clinical research evaluating the use of melatonin in COVID‑19 has shown various improved outcomes, including reduced hospitalization durations; however, the trials are small. Melatonin can alleviate mitochondrial dysfunction in COVID‑19, improve immune cell function and provide antioxidant properties. However, its therapeutic potential remains underexplored due to funding limitations and thus further investigations are required.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Department of Pathophysiology, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vasiliki Epameinondas Georgakopoulou
- Department of Pathophysiology, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX 78229, USA
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
32
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
33
|
Farahani E, Reinert LS, Narita R, Serrero MC, Skouboe MK, van der Horst D, Assil S, Zhang B, Iversen MB, Gutierrez E, Hazrati H, Johannsen M, Olagnier D, Kunze R, Denham M, Mogensen TH, Lappe M, Paludan SR. The HIF transcription network exerts innate antiviral activity in neurons and limits brain inflammation. Cell Rep 2024; 43:113792. [PMID: 38363679 PMCID: PMC10915869 DOI: 10.1016/j.celrep.2024.113792] [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: 08/07/2023] [Revised: 12/04/2023] [Accepted: 01/29/2024] [Indexed: 02/18/2024] Open
Abstract
Pattern recognition receptors (PRRs) induce host defense but can also induce exacerbated inflammatory responses. This raises the question of whether other mechanisms are also involved in early host defense. Using transcriptome analysis of disrupted transcripts in herpes simplex virus (HSV)-infected cells, we find that HSV infection disrupts the hypoxia-inducible factor (HIF) transcription network in neurons and epithelial cells. Importantly, HIF activation leads to control of HSV replication. Mechanistically, HIF activation induces autophagy, which is essential for antiviral activity. HSV-2 infection in vivo leads to hypoxia in CNS neurons, and mice with neuron-specific HIF1/2α deficiency exhibit elevated viral load and augmented PRR signaling and inflammatory gene expression in the CNS after HSV-2 infection. Data from human stem cell-derived neuron and microglia cultures show that HIF also exerts antiviral and inflammation-restricting activity in human CNS cells. Collectively, the HIF transcription factor system senses virus-induced hypoxic stress to induce cell-intrinsic antiviral responses and limit inflammation.
Collapse
Affiliation(s)
- Ensieh Farahani
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Line S Reinert
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Ryo Narita
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Manutea C Serrero
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Morten Kelder Skouboe
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Demi van der Horst
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Sonia Assil
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Baocun Zhang
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Marie B Iversen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Eugenio Gutierrez
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Hossein Hazrati
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Forensic Science, Aarhus University, Aarhus, Denmark
| | - Mogens Johannsen
- Department of Forensic Science, Aarhus University, Aarhus, Denmark
| | - David Olagnier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Mark Denham
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Lappe
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus, Denmark; CONNECT - Center for Clinical and Genomic Data, Aarhus University Hospital, Aarhus, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
34
|
Du L, Zhao J, Xie N, Xie H, Xu J, Bao X, Zhou Y, Liu H, Wu X, Hu X, He T, Xu S, Zheng Y. Protective effect and mechanism of Qingfei Paidu decoction on myocardial damage mediated by influenza viruses. Front Pharmacol 2024; 15:1309682. [PMID: 38476329 PMCID: PMC10927722 DOI: 10.3389/fphar.2024.1309682] [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: 10/08/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction: Significant attention has been paid to myocardial damage mediated by the single-stranded RNA virus. Qingfei Paidu decoction (QFPDD) has been proved to protect the damage caused by the influenza virus A/PR/8/1934 (PR8), but its specific mechanism is unclear. Methods: Molecular biological methods, together with network pharmacology, were used to analyze the effects and underlying mechanism of QFPDD treatment on PR8-induced myocardial damage to obtain insights into the treatment of COVID-19-mediated myocardial damage. Results: Increased apoptosis and subcellular damage were observed in myocardial cells of mice infected by PR8. QFPDD treatment significantly inhibited the apoptosis and subcellular damage induced by the PR8 virus. The inflammatory factors IFN-β, TNF-α, and IL-18 were statistically increased in the myocardia of the mice infected by PR8, and the increase in inflammatory factors was prevented by QFPDD treatment. Furthermore, the expression levels or phosphorylation of necroptosis-related proteins RIPK1, RIPK3, and MLKL were abnormally elevated in the group of infected mice, while QFPDD restored the levels or phosphorylation of these proteins. Our study demonstrated that HIF-1α is a key target of QFPDD in the treatment of influenza virus-mediated injury. The HIF-α level was significantly increased by PR8 infection. Both the knockdown of HIF-1α and treatment of the myocardial cell with QFPDD significantly reversed the increased inflammatory factors during infection. Overexpression of HIF-1α reversed the inhibition effects of QFPDD on cytokine expression. Meanwhile, seven compounds from QFPDD may target HIF-1α. Conclusion: QFPDD can ameliorate influenza virus-mediated myocardial damage by reducing the degree of cell necroptosis and apoptosis, inhibiting inflammatory response and the expression of HIF-1α. Thus, our results provide new insights into the treatment of respiratory virus-mediated myocardial damage.
Collapse
Affiliation(s)
- Lijuan Du
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
- Faculty of Physical Education, Ningbo University, Ningbo, Zhejiang, China
| | - Jing Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nanxi Xie
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huangze Xie
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Jiating Xu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Xiaoming Bao
- Department of Cardiology, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Yingsong Zhou
- Faculty of Physical Education, Ningbo University, Ningbo, Zhejiang, China
| | - Hui Liu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Hu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Tianyi He
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Shujun Xu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Cedillo-Barrón L, García-Cordero J, Visoso-Carvajal G, León-Juárez M. Viroporins Manipulate Cellular Powerhouses and Modulate Innate Immunity. Viruses 2024; 16:345. [PMID: 38543711 PMCID: PMC10974846 DOI: 10.3390/v16030345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 05/23/2024] Open
Abstract
Viruses have a wide repertoire of molecular strategies that focus on their replication or the facilitation of different stages of the viral cycle. One of these strategies is mediated by the activity of viroporins, which are multifunctional viral proteins that, upon oligomerization, exhibit ion channel properties with mild ion selectivity. Viroporins facilitate multiple processes, such as the regulation of immune response and inflammasome activation through the induction of pore formation in various cell organelle membranes to facilitate the escape of ions and the alteration of intracellular homeostasis. Viroporins target diverse membranes (such as the cellular membrane), endoplasmic reticulum, and mitochondria. Cumulative data regarding the importance of mitochondria function in multiple processes, such as cellular metabolism, energy production, calcium homeostasis, apoptosis, and mitophagy, have been reported. The direct or indirect interaction of viroporins with mitochondria and how this interaction affects the functioning of mitochondrial cells in the innate immunity of host cells against viruses remains unclear. A better understanding of the viroporin-mitochondria interactions will provide insights into their role in affecting host immune signaling through the mitochondria. Thus, in this review, we mainly focus on descriptions of viroporins and studies that have provided insights into the role of viroporins in hijacked mitochondria.
Collapse
Affiliation(s)
- Leticia Cedillo-Barrón
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN) Av., IPN # 2508 Col., San Pedro Zacatenco, Mexico City 07360, Mexico; (J.G.-C.); (G.V.-C.)
| | - Julio García-Cordero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN) Av., IPN # 2508 Col., San Pedro Zacatenco, Mexico City 07360, Mexico; (J.G.-C.); (G.V.-C.)
| | - Giovani Visoso-Carvajal
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN) Av., IPN # 2508 Col., San Pedro Zacatenco, Mexico City 07360, Mexico; (J.G.-C.); (G.V.-C.)
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón esq, Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Mexico City 11340, Mexico
| | - Moisés León-Juárez
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico;
| |
Collapse
|
36
|
May L, Chu CF, Zielinski CE. Single-Cell RNA Sequencing Reveals HIF1A as a Severity-Sensitive Immunological Scar in Circulating Monocytes of Convalescent Comorbidity-Free COVID-19 Patients. Cells 2024; 13:300. [PMID: 38391913 PMCID: PMC10886588 DOI: 10.3390/cells13040300] [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: 12/22/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is characterized by a wide range of clinical symptoms and a poorly predictable disease course. Although in-depth transcriptomic investigations of peripheral blood samples from COVID-19 patients have been performed, the detailed molecular mechanisms underlying an asymptomatic, mild or severe disease course, particularly in patients without relevant comorbidities, remain poorly understood. While previous studies have mainly focused on the cellular and molecular dissection of ongoing COVID-19, we set out to characterize transcriptomic immune cell dysregulation at the single-cell level at different time points in patients without comorbidities after disease resolution to identify signatures of different disease severities in convalescence. With single-cell RNA sequencing, we reveal a role for hypoxia-inducible factor 1-alpha (HIF1A) as a severity-sensitive long-term immunological scar in circulating monocytes of convalescent COVID-19 patients. Additionally, we show that circulating complexes formed by monocytes with either T cells or NK cells represent a characteristic cellular marker in convalescent COVID-19 patients irrespective of their preceding symptom severity. Together, these results provide cellular and molecular correlates of recovery from COVID-19 and could help in immune monitoring and in the design of new treatment strategies.
Collapse
Affiliation(s)
- Lilly May
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Chang-Feng Chu
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Christina E. Zielinski
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
- Department of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| |
Collapse
|
37
|
Móvio MI, de Almeida GWC, Martines IDGL, Barros de Lima G, Sasaki SD, Kihara AH, Poole E, Nevels M, Carlan da Silva MC. SARS-CoV-2 ORF8 as a Modulator of Cytokine Induction: Evidence and Search for Molecular Mechanisms. Viruses 2024; 16:161. [PMID: 38275971 PMCID: PMC10819295 DOI: 10.3390/v16010161] [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: 11/28/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/27/2024] Open
Abstract
Severe cases of SARS-CoV-2 infection are characterized by an immune response that leads to the overproduction of pro-inflammatory cytokines, resulting in lung damage, cardiovascular symptoms, hematologic symptoms, acute kidney injury and multiple organ failure that can lead to death. This remarkable increase in cytokines and other inflammatory molecules is primarily caused by viral proteins, and particular interest has been given to ORF8, a unique accessory protein specific to SARS-CoV-2. Despite plenty of research, the precise mechanisms by which ORF8 induces proinflammatory cytokines are not clear. Our investigations demonstrated that ORF8 augments production of IL-6 induced by Poly(I:C) in human embryonic kidney (HEK)-293 and monocyte-derived dendritic cells (mono-DCs). We discuss our findings and the multifaceted roles of ORF8 as a modulator of cytokine response, focusing on type I interferon and IL-6, a key component of the immune response to SARS-CoV-2. In addition, we explore the hypothesis that ORF8 may act through pattern recognition receptors of dsRNA such as TLRs.
Collapse
Affiliation(s)
- Marília Inês Móvio
- Laboratório de Neurogenética, Universidade Federal do ABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (M.I.M.)
| | - Giovana Waner Carneiro de Almeida
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do UFABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (G.W.C.d.A.); (G.B.d.L.); (S.D.S.)
| | - Isabella das Graças Lopes Martines
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do UFABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (G.W.C.d.A.); (G.B.d.L.); (S.D.S.)
| | - Gilmara Barros de Lima
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do UFABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (G.W.C.d.A.); (G.B.d.L.); (S.D.S.)
| | - Sergio Daishi Sasaki
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do UFABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (G.W.C.d.A.); (G.B.d.L.); (S.D.S.)
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (M.I.M.)
| | - Emma Poole
- Division of Virology, Department of Pathology, Cambridge University, Level 5, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Michael Nevels
- School of Biology, University of St Andrews, St Andrews KY16 9ST, UK;
| | - Maria Cristina Carlan da Silva
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do UFABC (UFABC), São Bernardo do Campo, São Paulo 09606-070, Brazil; (G.W.C.d.A.); (G.B.d.L.); (S.D.S.)
| |
Collapse
|
38
|
Sivagurunathan N, Calivarathan L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:431-448. [PMID: 37073650 DOI: 10.2174/1871527322666230418114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 04/20/2023]
Abstract
The pandemic of coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has become a global concern as it leads to a spectrum of mild to severe symptoms and increases death tolls around the world. Severe COVID-19 results in acute respiratory distress syndrome, hypoxia, and multi- organ dysfunction. However, the long-term effects of post-COVID-19 infection are still unknown. Based on the emerging evidence, there is a high possibility that COVID-19 infection accelerates premature neuronal aging and increases the risk of age-related neurodegenerative diseases in mild to severely infected patients during the post-COVID period. Several studies correlate COVID-19 infection with neuronal effects, though the mechanism through which they contribute to the aggravation of neuroinflammation and neurodegeneration is still under investigation. SARS-CoV-2 predominantly targets pulmonary tissues and interferes with gas exchange, leading to systemic hypoxia. The neurons in the brain require a constant supply of oxygen for their proper functioning, suggesting that they are more vulnerable to any alteration in oxygen saturation level that results in neuronal injury with or without neuroinflammation. We hypothesize that hypoxia is one of the major clinical manifestations of severe SARS-CoV-2 infection; it directly or indirectly contributes to premature neuronal aging, neuroinflammation, and neurodegeneration by altering the expression of various genes responsible for the survival of the cells. This review focuses on the interplay between COVID-19 infection, hypoxia, premature neuronal aging, and neurodegenerative diseases and provides a novel insight into the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| |
Collapse
|
39
|
Dhaliwal M, Muthu V, Sharma A, Raj K, Rudramurthy SM, Agarwal R, Kaur H, Rawat A, Singh S, Chakrabarti A. Immune and metabolic perturbations in COVID-19-associated pulmonary mucormycosis: A transcriptome analysis of innate immune cells. Mycoses 2024; 67:e13679. [PMID: 38214399 DOI: 10.1111/myc.13679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/23/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND AND OBJECTIVES The mechanisms underlying COVID-19-associated pulmonary mucormycosis (CAPM) remain unclear. We use a transcriptomic analysis of the innate immune cells to investigate the host immune and metabolic response pathways in patients with CAPM. PATIENTS AND METHODS We enrolled subjects with CAPM (n = 5), pulmonary mucormycosis (PM) without COVID-19 (n = 5), COVID-19 (without mucormycosis, n = 5), healthy controls (n = 5) without comorbid illness and negative for SARS-CoV-2. Peripheral blood samples from cases were collected before initiating antifungal therapy, and neutrophils and monocytes were isolated. RNA sequencing was performed using Illumina HiSeqX from monocytes and neutrophils. Raw reads were aligned with HISAT-2 pipeline and DESeq2 was used for differential gene expression. Gene ontology (GO) and metabolic pathway analysis were performed using Shiny GO application and R packages (ggplot2, Pathview). RESULTS The derangement of core immune and metabolic responses in CAPM patients was noted. Pattern recognition receptors, dectin-2, MCL, FcRγ receptors and CLEC-2, were upregulated, but signalling pathways such as JAK-STAT, IL-17 and CARD-9 were downregulated; mTOR and MAP-kinase signalling were elevated in monocytes from CAPM patients. The complement receptors, NETosis, and pro-inflammatory responses, such as S100A8/A9, lipocalin and MMP9, were elevated. The major metabolic pathways of glucose metabolism-glycolysis/gluconeogenesis, pentose phosphate pathway, HIF signalling and iron metabolism-ferroptosis were also upregulated in CAPM. CONCLUSIONS We identified significant alterations in the metabolic pathways possibly leading to cellular iron overload and a hyperglycaemic state. Immune responses revealed altered recognition, signalling, effector functions and a pro-inflammatory state in monocytes and neutrophils from CAPM patients.
Collapse
Affiliation(s)
- Manpreet Dhaliwal
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Valliappan Muthu
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunima Sharma
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Khem Raj
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harsimran Kaur
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
40
|
Cojocaru E, Cojocaru T, Pînzariu GM, Vasiliu I, Armașu I, Cojocaru C. Perspectives on Post-COVID-19 Pulmonary Fibrosis Treatment. J Pers Med 2023; 14:51. [PMID: 38248752 PMCID: PMC10817460 DOI: 10.3390/jpm14010051] [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: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Pulmonary fibrosis, a critical outcome of chronic inflammatory diseases, has gained prominence in the context of post-coronavirus (post-COVID-19) complications. This review delves into the multifaceted landscape of post-COVID-19 pulmonary fibrosis, elucidating the intricate molecular mechanisms underlying its pathogenesis and highlighting promising therapeutic avenues. Examining the aftermath of severe acute respiratory syndrome-2 (SARS-CoV-2) infection, the review reveals key signaling pathways implicated in the fibrotic cascade. Drawing parallels with previous coronavirus outbreaks enhances our understanding of the distinctive features of post-COVID-19 fibrosis. Antifibrotic drugs, like pirfenidone and nintedanib, take center stage; their mechanisms of action and potential applications in post-COVID-19 cases are thoroughly explored. Beyond the established treatments, this review investigates emerging therapeutic modalities, including anti-interleukin agents, immunosuppressants, and experimental compounds, like buloxybutide, saracatinib, sirolimus, and resveratrol. Emphasizing the critical importance of early intervention, this review highlights the dynamic nature of post-COVID-19 pulmonary fibrosis research. In conclusion, the synthesis of current knowledge offers a foundation for advancing our approaches to the prevention and treatment of these consequential sequelae of COVID-19.
Collapse
Affiliation(s)
- Elena Cojocaru
- Morpho-Functional Sciences II Department, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (E.C.); (I.V.)
| | - Tudor Cojocaru
- Faculty of Medicine, University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (G.M.P.); (I.A.)
| | - Giulia Mihaela Pînzariu
- Faculty of Medicine, University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (G.M.P.); (I.A.)
| | - Ioana Vasiliu
- Morpho-Functional Sciences II Department, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (E.C.); (I.V.)
| | - Ioana Armașu
- Faculty of Medicine, University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (G.M.P.); (I.A.)
| | - Cristian Cojocaru
- Medical III Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
41
|
Rago V, Bossio S, Lofaro D, Perri A, Di Agostino S. New Insights into the Link between SARS-CoV-2 Infection and Renal Cancer. Life (Basel) 2023; 14:52. [PMID: 38255667 PMCID: PMC10817602 DOI: 10.3390/life14010052] [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] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Cancer has been described as a risk factor for greater susceptibility to SARS-CoV-2 infection and severe COVID-19, mainly for patients with metastatic disease. Conversely, to that reported for most solid and hematological malignancies, the few available clinical studies reported that the infection did not increase the risk of death in renal cancer patients. The expression on proximal tubular renal cells of the key players in cellular viral uptake, ACE2, TMPRSS2, and NRP1, seems to be the mechanism for the direct kidney injury seen in patients with COVID-19. Interestingly, data from The Cancer Genome Atlas and experimental analyses on various renal cancer cell lines demonstrated that the above-reported receptors/cofactors are maintained by renal cancer cells. However, whether SARS-CoV-2 infection directly kills renal cancer cells or generates enhanced immunogenicity is a question worth investigating. In addition, some researchers have further addressed the topic by studying the expression and prognostic significance of gene signatures related to SARS-CoV-2 infection in renal cancer patients. The emerging data highlights the importance of better understanding the existence of a link between renal cancer and COVID-19 since it could lead to the identification of new prognostic factors and the development of new therapeutic targets in the management of renal cancer patients.
Collapse
Affiliation(s)
- Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Sabrina Bossio
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Danilo Lofaro
- de-Health Lab, Department of Mechanical, Energy, Management Engineering, University of Calabria, 87036 Rende, Italy;
| | - Anna Perri
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Silvia Di Agostino
- Department of Health Sciences, Magna Græcia University of Catanzaro, 88100 Catanzaro, Italy;
| |
Collapse
|
42
|
Verma M, Rawat N, Rani R, Singh M, Choudhary A, Abbasi S, Kumar M, Kumar S, Tanwar A, Misir BR, Khanna S, Agrawal A, Faruq M, Rai S, Tripathi R, Kumar A, Pujani M, Bhojani M, Pandey AK, Nesari T, Prasher B. Adhatoda vasica and Tinospora cordifolia extracts ameliorate clinical and molecular markers in mild COVID-19 patients: a randomized open-label three-armed study. Eur J Med Res 2023; 28:556. [PMID: 38049897 PMCID: PMC10696694 DOI: 10.1186/s40001-023-01507-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 11/04/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infections caused mild-to-moderate illness. However, a sizable portion of infected people experience a rapid progression of hyper-inflammatory and hypoxic respiratory illness that necessitates an effective and safer remedy to combat COVID-19. METHODS A total of 150 COVID-19-positive patients with no to mild symptoms, between the age groups 19-65 years were enrolled in this randomized, open-labeled three-armed clinical trial. Among them, 136 patients completed the study with RT-PCR negative reports. The patients received herbal drugs orally (Group A (Adhatoda vasica; AV; 500 mg; n = 50); Group B (Tinospora cordifolia; TC; 500 mg; n = 43), and Group C (AV + TC; 250 mg each; n = 43)) for 14 days. Clinical symptoms, vital parameters, and viral clearance were taken as primary outcomes, and biochemical, hematological parameters, cytokines, and biomarkers were evaluated at three time points as secondary outcomes. RESULTS We found that the mean viral clearance time was 13.92 days (95% confidence interval [CI] 12.85-14.99) in Group A, 13.44 days (95% confidence interval [CI] 12.14-14.74) in Group B, and 11.86 days (95% confidence interval [CI] 10.62-13.11) days in Group C. Over a period of 14 days, the mean temperature in Groups A, and B significantly decreased linearly. In Group A, during the trial period, eosinophils, and PT/INR increased significantly, while monocytes, SGOT, globulin, serum ferritin, and HIF-1α, a marker of hypoxia reduced significantly. On the other hand, in Group B hsCRP decreased at mid-treatment. Eosinophil levels increased in Group C during the treatment, while MCP-3 levels were significantly reduced. CONCLUSIONS All the patients of the three-armed interventions recovered from COVID-19 and none of them reported any adverse effects from the drugs. Group C patients (AV + TC) resulted in a quicker viral clearance as compared to the other two groups. We provide the first clinical report of AV herbal extract acting as a modifier of HIF-1α in COVID-19 patients along with a reduction in levels of ferritin, VEGF, and PT/INR as the markers of hypoxia, inflammation, and thrombosis highlighting the potential use in progression stages, whereas the TC group showed immunomodulatory effects. Trial registration Clinical Trials Database -India (ICMR-NIMS), CTRI/2020/09/028043. Registered 24th September 2020, https://www.ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=47443&EncHid=&modid=&compid=%27,%2747443det%27.
Collapse
Grants
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
- GAP-0183 Ministry of AYUSH, Government of India
Collapse
Affiliation(s)
- Mukta Verma
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Neha Rawat
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Ritu Rani
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Manju Singh
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Aditi Choudhary
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sarfaraz Abbasi
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Manish Kumar
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sachin Kumar
- ESIC Medical College and Hospital, Faridabad, Haryana, India
| | - Ankur Tanwar
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- All India Institute of Ayurveda, New Delhi, India
| | - Bishnu Raman Misir
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Sangeeta Khanna
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Anurag Agrawal
- Trivedi School of Biosciences, Ashoka University, Sonipat, Haryana, India
| | - Mohammed Faruq
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India
| | - Shalini Rai
- All India Institute of Ayurveda, New Delhi, India
| | | | - Anil Kumar
- All India Institute of Ayurveda, New Delhi, India
| | - Mukta Pujani
- ESIC Medical College and Hospital, Faridabad, Haryana, India
| | | | | | | | - Bhavana Prasher
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, CSIR-Institute of Genomics & Integrative Biology, Delhi, India.
- CSIR-Institute of Genomics & Integrative Biology, Delhi, India.
| |
Collapse
|
43
|
Ma J, Chen X, Xue R, Wang F, Dong J, Tao N, Qin Z. Cinnamaldehyde inhibits cytokine storms induced by the ORF3a protein of SARS-CoV-2 via ROS-elimination in activated T cells. Phytother Res 2023; 37:6006-6020. [PMID: 37726983 DOI: 10.1002/ptr.8016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/04/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023]
Abstract
Cytokine storms are the cause of complications in patients with severe COVID-19, and it becomes the target of therapy. Several natural compounds were selected to screen the inhibitory effect on T-cell proliferation by Fluorescence-Activated Cell Sorting (FACS) and cytokine production by enzyme-linked immunosorbent assay (ELISA). Open reading frame 3a (ORF3a) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the specific T-cell activation model in vivo and in vitro. The coculture system included the macrophage cell line RAW264.7 and splenocytes. Reactive oxygen species (ROS) levels and glycolysis in T cells were evaluated. Cinnamaldehyde effectively inhibits cytokine storms both in vitro and in vivo. It decreased inflammatory cytokine (such as IFN-γ, TNF-α, IL-6, and IL-2) production by murine peripheral blood cells upon direct stimulation with ConA, after immunization with the MHV-A59 virus or ORF3a peptide from SARS-CoV-2. Cinnamaldehyde restored the percentage of T cells, which was originally decreased in the peripheral blood and splenocytes of ORF3a-immunized mice. In a coculture system, cinnamaldehyde reduced the secretion of inflammatory cytokines from macrophages in a T-cell dependent manner. Furthermore, cinnamaldehyde decreased the ROS level in activated T cells, which in turn reduced glycolysis and the activation of T cells. Cinnamaldehyde can be used as a candidate molecule for COVID-19.
Collapse
Affiliation(s)
- Jing Ma
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xu Chen
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Ning Tao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
44
|
Hotani T, Nakagawa K, Tsukamoto T, Mizuguchi H, Sakurai F. Involvement of Protein Kinase R in Double-Stranded RNA-Induced Proteasomal Degradation of Hypoxia Inducible Factor-1α. Inflammation 2023; 46:2332-2342. [PMID: 37615898 PMCID: PMC10673737 DOI: 10.1007/s10753-023-01881-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023]
Abstract
Hypoxia inducible factor-1α (HIF-1α) is a crucial therapeutic target in various diseases, including cancer and fibrosis. We previously demonstrated that transfection with double-stranded RNA (dsRNA), including polyI:C and the dsRNA genome of mammalian orthoreovirus, resulted in significant reduction in HIF-1α protein levels in cultured cells; however, it remained to be elucidated how dsRNA induced down-regulation of HIF-1α protein levels. In this study, we examined the mechanism of dsRNA-mediated down-regulation of HIF-1α protein levels. We found that among the various cellular factors involved in dsRNA-mediated innate immunity, knockdown and knockout of protein kinase R (PKR) significantly restored HIF-1α protein levels in dsRNA-transfected cells, indicating that PKR was involved in dsRNA-mediated down-regulation of HIF-1α. Proteasome inhibitors significantly restored the HIF-1α protein levels in dsRNA-transfected cells. Ubiquitination levels of HIF-1α were increased by transfection with dsRNA. These findings indicated that degradation of HIF-1α in a ubiquitin-proteasome pathway was promoted in a PKR-dependent manner following dsRNA transfection. Expression of not only HIF-1α but also several proteins, including CDK4 and HER2, was down-regulated following dsRNA transfection. These data provide important clues for elucidation of the mechanism of dsRNA-mediated cellular toxicity, as well as for therapeutic application of dsRNA.
Collapse
Affiliation(s)
- Takuma Hotani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kanako Nakagawa
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomohito Tsukamoto
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- The Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Laboratory of Functional Organoid for Drug Discovery, Center for Drug Discovery Resources Research, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito, Asagi, Ibaraki, Osaka, 567-0085, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, 565-0871, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
45
|
Zhang Z, Zhang N, Lu X, Zhou M, Yan X, Gu W, Yang J, Zhang Q, Zhang C, Gong Y, Jia M, Zhang X, Ning P, Liu M, Li X, Shi X, Liu W, Gao GF, Ning G, Wang J, Bi Y. Anti-infection effects of heparin on SARS-CoV-2 in a diabetic mouse model. Zool Res 2023; 44:1003-1014. [PMID: 37759335 PMCID: PMC10802103 DOI: 10.24272/j.issn.2095-8137.2023.108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in more severe syndromes and poorer outcomes in patients with diabetes and obesity. However, the precise mechanisms responsible for the combined impact of corona virus disease 2019 (COVID-19) and diabetes have not yet been elucidated, and effective treatment options for SARS-CoV-2-infected diabetic patients remain limited. To investigate the disease pathogenesis, K18-hACE2 transgenic (hACE2 Tg) mice with a leptin receptor deficiency (hACE2-Lepr -/-) or high-fat diet (hACE2-HFD) background were generated. The two mouse models were intranasally infected with a 5×10 5 median tissue culture infectious dose (TCID 50) of SARS-CoV-2, with serum and lung tissue samples collected at 3 days post-infection. The hACE2-Lepr -/- mice were then administered a combination of low-molecular-weight heparin (LMWH) (1 mg/kg or 5 mg/kg) and insulin via subcutaneous injection prior to intranasal infection with 1×10 4 TCID 50 of SARS-CoV-2. Daily drug administration continued until the euthanasia of the mice. Analyses of viral RNA loads, histopathological changes in lung tissue, and inflammation factors were conducted. Results demonstrated similar SARS-CoV-2 susceptibility in hACE2 Tg mice under both lean (chow diet) and obese (HFD) conditions. However, compared to the hACE2-Lepr +/+ mice, hACE2-Lepr -/- mice exhibited more severe lung injury, enhanced expression of inflammatory cytokines and hypoxia-inducible factor-1α, and increased apoptosis. Moreover, combined LMWH and insulin treatment effectively reduced disease progression and severity, attenuated lung pathological changes, and mitigated inflammatory responses. In conclusion, pre-existing diabetes can lead to more severe lung damage upon SARS-CoV-2 infection, and LMWH may be a valuable therapeutic approach for managing COVID-19 patients with diabetes.
Collapse
Affiliation(s)
- Zhongyun Zhang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Ning Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxiang Yan
- Department of Cardiology, Institute of Cardiovascular Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jingru Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qin Zhang
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Cheng Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Yuhuan Gong
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mingjun Jia
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Ning
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Liu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaoyan Li
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaomeng Shi
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang Ning
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China. E-mail:
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- University of Chinese Academy of Sciences, Beijing 100049, China. E-mail:
| |
Collapse
|
46
|
Jeong K, Kim Y, Jeon J, Kim K. Subtyping of COVID-19 samples based on cell-cell interaction in single cell transcriptomes. Sci Rep 2023; 13:19629. [PMID: 37949890 PMCID: PMC10638268 DOI: 10.1038/s41598-023-46350-2] [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: 08/11/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
In single-cell transcriptome analysis, numerous biomarkers related to COVID-19 severity, including cell subtypes, genes, and pathways, have been identified. Nevertheless, most studies have focused on severity groups based on clinical features, neglecting immunological heterogeneity within the same severity level. In this study, we employed sample-level clustering using cell-cell interaction scores to investigate patient heterogeneity and uncover novel subtypes. The clustering results were validated using external datasets, demonstrating superior reproducibility and purity compared to gene expression- or gene set enrichment-based clustering. Furthermore, the cell-cell interaction score-based clusters exhibited a strong correlation with the WHO ordinal severity score based on clinical characteristics. By characterizing the identified subtypes through known COVID-19 severity-associated biomarkers, we discovered a "Severe-like moderate" subtype. This subtype displayed clinical features akin to moderate cases; however, molecular features, such as gene expression and cell-cell interactions, resembled those of severe cases. Notably, all patients who progressed from moderate to severe belonged to this subtype, underscoring the significance of cell-cell interactions in COVID-19 patient heterogeneity and severity.
Collapse
Affiliation(s)
- Kyeonghun Jeong
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yooeun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaemin Jeon
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
- Department of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
47
|
Amadoro G, Latina V, Stigliano E, Micera A. COVID-19 and Alzheimer's Disease Share Common Neurological and Ophthalmological Manifestations: A Bidirectional Risk in the Post-Pandemic Future. Cells 2023; 12:2601. [PMID: 37998336 PMCID: PMC10670749 DOI: 10.3390/cells12222601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
A growing body of evidence indicates that a neuropathological cross-talk takes place between the coronavirus disease 2019 (COVID-19) -the pandemic severe pneumonia that has had a tremendous impact on the global economy and health since three years after its outbreak in December 2019- and Alzheimer's Disease (AD), the leading cause of dementia among human beings, reaching 139 million by the year 2050. Even though COVID-19 is a primary respiratory disease, its causative agent, the so-called Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), is also endowed with high neuro-invasive potential (Neurocovid). The neurological complications of COVID-19, resulting from the direct viral entry into the Central Nervous System (CNS) and/or indirect systemic inflammation and dysregulated activation of immune response, encompass memory decline and anosmia which are typically associated with AD symptomatology. In addition, patients diagnosed with AD are more vulnerable to SARS-CoV-2 infection and are inclined to more severe clinical outcomes. In the present review, we better elucidate the intimate connection between COVID-19 and AD by summarizing the involved risk factors/targets and the underlying biological mechanisms shared by these two disorders with a particular focus on the Angiotensin-Converting Enzyme 2 (ACE2) receptor, APOlipoprotein E (APOE), aging, neuroinflammation and cellular pathways associated with the Amyloid Precursor Protein (APP)/Amyloid beta (Aβ) and tau neuropathologies. Finally, the involvement of ophthalmological manifestations, including vitreo-retinal abnormalities and visual deficits, in both COVID-19 and AD are also discussed. Understanding the common physiopathological aspects linking COVID-19 and AD will pave the way to novel management and diagnostic/therapeutic approaches to cope with them in the post-pandemic future.
Collapse
Affiliation(s)
- Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy;
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Valentina Latina
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy;
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Egidio Stigliano
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
| |
Collapse
|
48
|
Wigner-Jeziorska P, Janik-Karpińska E, Niwald M, Saluk J, Miller E. Effect of SARS-CoV-2 Infection and BNT162b2 Vaccination on the mRNA Expression of Genes Associated with Angiogenesis. Int J Mol Sci 2023; 24:16094. [PMID: 38003287 PMCID: PMC10671623 DOI: 10.3390/ijms242216094] [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: 09/11/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), discovered in December 2019 in Wuhan, China, caused the coronavirus disease 2019 (COVID-19). Due to the rate of spread of this virus, the World Health Organization, in March 2020, recognised COVID-19 as a worldwide pandemic. The disease is multisystemic with varying degrees of severity. Unfortunately, despite intensive research, the molecular changes caused by SARS-CoV-2 remain unclear. Mechanisms affected by the virus infection include endothelial dysfunction and angiogenesis. Similarly, the vaccines developed so far affect the process of angiogenesis, contributing to the development of undesirable effects on part of the cardiovascular system. The presented research aimed to investigate the impact of the SARS-CoV-2 infection and the Pfizer Comirnaty vaccine (BNT162b2) on the molecular aspect of angiogenesis. We found that convalescents vaccinated with one dose of BNT162b2 were characterised by higher MMP-7 (metalloproteinases 7) expression than non-vaccinated convalescents and healthy volunteers vaccinated with one dose of BNT162b2. Moreover, non-vaccinated convalescents showed increased mRNA expression of ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif 1) compared to healthy volunteers vaccinated with one dose of BNT162b2. In addition, we showed significant sex differences in the expression of MMP-7. In conclusion, the results of our study suggest a significant impact of SARS-CoV-2 infection and vaccination on the course of angiogenesis at the molecular level.
Collapse
Affiliation(s)
- Paulina Wigner-Jeziorska
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.-J.); (J.S.)
| | - Edyta Janik-Karpińska
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Marta Niwald
- Department of Neurological Rehabilitation, Medical University of Lodz, 90-136 Lodz, Poland;
| | - Joanna Saluk
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.-J.); (J.S.)
| | - Elżbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, 90-136 Lodz, Poland;
| |
Collapse
|
49
|
Shen Y, Peng A, Chu X, Ji Z, Li M, Bai S, Wei Z, Liang H, Zhao X, Xue G. Association of seizures with COVID-19 infection in underage during the pandemic: A systematic review and meta-analysis. Epilepsy Res 2023; 197:107223. [PMID: 37801748 DOI: 10.1016/j.eplepsyres.2023.107223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE Coronavirus disease 2019 (COVID-19) is a contagious infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has caused worldwide transmission. The aim of this systematic review and meta-analysis was to investigate the morbidity and characteristics of seizures after SARS-CoV-2 infection in underage (≤18 y) and to provide valuable reference material for subsequent clinical treatment. METHODS PubMed/MEDLINE, Cochrane, and EMBASE databases were searched up to 10th May 2023. We utilized the search strategy of medical subject headings combined with entry terms to search all related literatures. RESULTS The meta-analysis was performed according to PRISMA reporting guidelines. Risk of bias was assessed using the Newcastle-Ottawa Scale (NOS) and the Agency for Healthcare Research and Quality (AHRQ). A total of 12 articles were selected, including 4153 subjects and 333 seizure-prone minor patients with COVID-19. The morbidity of seizures after SARS-CoV-2 infection in immature patients was approximately 8.2 (95 % CI, 4.7%-12.4 %). By subgroup analysis, we know that the morbidity of male, Americas, with fever and first occurrence of seizures were 4.2% (95 % CI, 0.4-10.5 %), 4.6 % (95 % CI, 0.4 %-11.7 %), 5.4 % (95 % CI, 2.6 %-9.1 %) and 3.7 % (95 % CI, 0.7 %-8.2 %), respectively. Generalized seizures are the main type of seizures (80.6 %). CONCLUSIONS Seizures can be caused by SARS-CoV-2 infection in underage groups, with a pooled morbidity of 8.2% and a higher morbidity in females, in African regions, in febrile groups and during 2022-2023. In addition, generalized seizures being the predominant seizure type.
Collapse
Affiliation(s)
- Yingjie Shen
- School of Yuncheng Clinical Medicine, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China; Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Anqi Peng
- School of Yuncheng Clinical Medicine, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China
| | - Xuehong Chu
- School of Yuncheng Clinical Medicine, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China
| | - Zhongjie Ji
- School of Yuncheng Clinical Medicine, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China
| | - Mengkai Li
- Department of neurosurgery, Yuncheng Central Hospital, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China
| | - Sha Bai
- Capital University of Economics and Business, Beijing, China
| | - Zhongjun Wei
- Department of neurosurgery, Yuncheng Central Hospital, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Xujing Zhao
- Department of Pediatrics, Yuncheng Pediatrics Hospital, No. 330, Hedong East Street, Yuncheng 044000, Shanxi Province, China.
| | - Guoqiang Xue
- Department of neurosurgery, Yuncheng Central Hospital, Shanxi Medical University, No. 3690, Hedong East Street, Yuncheng 044000, Shanxi Province, China.
| |
Collapse
|
50
|
Briassoulis G, Briassoulis P, Ilia S, Miliaraki M, Briassouli E. The Anti-Oxidative, Anti-Inflammatory, Anti-Apoptotic, and Anti-Necroptotic Role of Zinc in COVID-19 and Sepsis. Antioxidants (Basel) 2023; 12:1942. [PMID: 38001795 PMCID: PMC10669546 DOI: 10.3390/antiox12111942] [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: 09/23/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Zinc is a structural component of proteins, functions as a catalytic co-factor in DNA synthesis and transcription of hundreds of enzymes, and has a regulatory role in protein-DNA interactions of zinc-finger proteins. For many years, zinc has been acknowledged for its anti-oxidative and anti-inflammatory functions. Furthermore, zinc is a potent inhibitor of caspases-3, -7, and -8, modulating the caspase-controlled apoptosis and necroptosis. In recent years, the immunomodulatory role of zinc in sepsis and COVID-19 has been investigated. Both sepsis and COVID-19 are related to various regulated cell death (RCD) pathways, including apoptosis and necroptosis. Lack of zinc may have a negative effect on many immune functions, such as oxidative burst, cytokine production, chemotaxis, degranulation, phagocytosis, and RCD. While plasma zinc concentrations decline swiftly during both sepsis and COVID-19, this reduction is primarily attributed to a redistribution process associated with the inflammatory response. In this response, hepatic metallothionein production increases in reaction to cytokine release, which is linked to inflammation, and this protein effectively captures and stores zinc in the liver. Multiple regulatory mechanisms come into play, influencing the uptake of zinc, the binding of zinc to blood albumin and red blood cells, as well as the buffering and modulation of cytosolic zinc levels. Decreased zinc levels are associated with increasing severity of organ dysfunction, prolonged hospital stay and increased mortality in septic and COVID-19 patients. Results of recent studies focusing on these topics are summarized and discussed in this narrative review. Existing evidence currently does not support pharmacological zinc supplementation in patients with sepsis or COVID-19. Complementation and repletion should follow current guidelines for micronutrients in critically ill patients. Further research investigating the pharmacological mechanism of zinc in programmed cell death caused by invasive infections and its therapeutic potential in sepsis and COVID-19 could be worthwhile.
Collapse
Affiliation(s)
- George Briassoulis
- Postgraduate Program “Emergency and Intensive Care in Children, Adolescents, and Young Adults”, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Panagiotis Briassoulis
- Second Department of Anesthesiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Stavroula Ilia
- Postgraduate Program “Emergency and Intensive Care in Children, Adolescents, and Young Adults”, School of Medicine, University of Crete, 71003 Heraklion, Greece;
- Paediatric Intensive Care Unit, University Hospital, School of Medicine, University of Crete, 71110 Heraklion, Greece;
| | - Marianna Miliaraki
- Paediatric Intensive Care Unit, University Hospital, School of Medicine, University of Crete, 71110 Heraklion, Greece;
| | - Efrossini Briassouli
- Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|