51
|
Wang H, Cong L, Yin X, Zhang N, Zhu M, Sun T, Fan J, Xue F, Fan X, Gong Y. The Apelin-APJ axis alleviates LPS-induced pulmonary fibrosis and endothelial mesenchymal transformation in mice by promoting Angiotensin-Converting Enzyme 2. Cell Signal 2022; 98:110418. [PMID: 35882286 DOI: 10.1016/j.cellsig.2022.110418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022]
Abstract
Fibrotic alterations resulting from abnormal tissue repair after lung injury are responsible for the high mortality observed after acute respiratory distress syndrome. Therefore, the prevention and treatment of pulmonary fibrosis has been widely concerned. The Apelin-APJ axis plays an important role in the prevention and treatment of respiratory diseases and organ fibrosis. However, its underlying mechanism remains to be further studied. The aim of this study was to investigate whether the anti-pulmonary fibrosis effect of apelin-APJ axis is related to the activation of angiotensin-converting Enzyme 2 (ACE2). Here, we found that exogenous activation of the Apelin-APJ axis alleviates lipopolysaccharide (LPS)-induced pulmonary fibrosis in mice. In vitro studies revealed that Apelin-13 inhibited LPS-induced endothelial mesenchymal transition in lung microvascular endothelial cells, whereas [Ala13]-Apelin-13 (Apelin-APJ axis inhibitor) accelerated LPS-induced endothelial interstitial transformation in lung microvascular endothelial cells. Notably, angiotensin-converting enzyme 2 (ACE2) inhibitor blocks the beneficial effect of the Apelin-APJ axis activation on LPS-induced pulmonary fibrosis. This finding suggests that the Apelin-APJ axis inhibits pulmonary fibrosis by activating ACE2. Simultaneously, accumulating evidence suggests that ubiquitination may contribute to pulmonary fibrosis. Our study found that LPS increased the ubiquitination of ACE2 protein, whereas Apelin-13 inhibited it. In conclusion, exogenous activation of the Apelin-APJ axis improves LPS-induced pulmonary fibrosis in mice and may be a viable therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Linjing Cong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Xianghong Yin
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Nan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Min Zhu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Tingting Sun
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Feng Xue
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China.
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences,Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
52
|
Yang Z, Zhou L, Ge H, Shen W, Shan L. Identification of autophagy-related biomarkers in patients with pulmonary arterial hypertension based on bioinformatics analysis. Open Med (Wars) 2022; 17:1148-1157. [PMID: 35859795 PMCID: PMC9263897 DOI: 10.1515/med-2022-0497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Autophagy participates in the regulation of pulmonary arterial hypertension (PAH). However, the role of autophagy-related genes (ARGs) in the pathogenesis of the PAH is still unclear. This study aimed to identify the ARGs in PAH via bioinformatics analysis. A microarray dataset (GSE113439) was downloaded from the Gene Expression Omnibus database to identify differentially expressed ARGs (DEARGs). Protein–protein interactions network, gene ontology, and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to screen hub genes and the underlying molecular mechanisms of PAH. Finally, the mRNA expression of the hub genes was validated using the GSE53408 dataset. Twenty-six DEARGs were identified, all of which were upregulated. Enrichment analyses revealed that these DEARGs were mainly enriched in the nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathway, PI3K-Akt signaling pathway, response to hypoxia, response to nutrient levels, and autophagy. Among these hub genes, the mRNA expression levels of HSP90AA1, HIF1A, MET, IGF1, LRRK2, CLTC, DNM1L, MDM2, RICTOR, and ROCK2 were significantly upregulated in PAH patients than in healthy individuals. Ten hub DEARGs were identified and may participate in the pathogenesis of the PAH via the regulation of autophagy. The present study may provide novel therapeutic targets for PAH prevention and treatment and expand our understanding of PAH.
Collapse
Affiliation(s)
- Zhisong Yang
- Department of Emergency, Daqing Longnan Hospital, Daqing, Heilongjiang 163453, China
| | - Li Zhou
- Department of Emergency, Daqing Longnan Hospital, Daqing, Heilongjiang 163453, China
| | - Haiyan Ge
- Department of Respiratory Medicine, Shanghai Huadong Hospital, Shanghai 200040, China
| | - Weimin Shen
- Department of Respiratory Medicine, Shanghai Huadong Hospital, Shanghai 200040, China
| | - Lin Shan
- Department of Respiratory Medicine, Shanghai Huadong Hospital, Shanghai 200040, China
| |
Collapse
|
53
|
Salt-inducible kinases: new players in pulmonary arterial hypertension? Trends Pharmacol Sci 2022; 43:806-819. [PMID: 35851157 DOI: 10.1016/j.tips.2022.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/13/2022]
Abstract
Salt-inducible kinases (SIKs) are serine/threonine kinases belonging to the AMP-activated protein kinase (AMPK) family. Accumulating evidence indicates that SIKs phosphorylate multiple targets, including histone deacetylases (HDACs) and cAMP response element-binding protein (CREB)-regulated transcriptional coactivators (CRTCs), to coordinate signaling pathways implicated in metabolism, cell growth, proliferation, apoptosis, and inflammation. These pathways downstream of SIKs are altered not only in pathologies like cancer, systemic hypertension, and inflammatory diseases, but also in pulmonary arterial hypertension (PAH), a multifactorial disease characterized by pulmonary vasoconstriction, inflammation and remodeling of pulmonary arteries owing to endothelial dysfunction and aberrant proliferation of smooth muscle cells (SMCs). In this opinion article, we present evidence of SIKs as modulators of key signaling pathways involved in PAH pathophysiology and discuss the potential of SIKs as therapeutic targets for PAH, emphasizing the need for deeper molecular insights on PAH.
Collapse
|
54
|
Flores K, Siques P, Brito J, Arribas SM. AMPK and the Challenge of Treating Hypoxic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms23116205. [PMID: 35682884 PMCID: PMC9181235 DOI: 10.3390/ijms23116205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary artery pressure produced by vasoconstriction and hyperproliferative remodeling of the pulmonary artery and subsequent right ventricular hypertrophy (RVH). The search for therapeutic targets for cardiovascular pathophysiology has extended in many directions. However, studies focused on mitigating high-altitude pulmonary hypertension (HAPH) have been rare. Because AMP-activated protein kinase (AMPK) is involved in cardiovascular and metabolic pathology, AMPK is often studied as a potential therapeutic target. AMPK is best characterized as a sensor of cellular energy that can also restore cellular metabolic homeostasis. However, AMPK has been implicated in other pathways with vasculoprotective effects. Notably, cellular metabolic stress increases the intracellular ADP/ATP or AMP/ATP ratio, and AMPK activation restores ATP levels by activating energy-producing catabolic pathways and inhibiting energy-consuming anabolic pathways, such as cell growth and proliferation pathways, promoting cardiovascular protection. Thus, AMPK activation plays an important role in antiproliferative, antihypertrophic and antioxidant pathways in the pulmonary artery in HPH. However, AMPK plays contradictory roles in promoting HPH development. This review describes the main findings related to AMPK participation in HPH and its potential as a therapeutic target. It also extrapolates known AMPK functions to discuss the less-studied HAPH context.
Collapse
Affiliation(s)
- Karen Flores
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
- Correspondence: ; Tel.: +56-572526392
| | - Patricia Siques
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Silvia M. Arribas
- Department of Physiology, University Autonoma of Madrid, 28049 Madrid, Spain;
| |
Collapse
|
55
|
Hasanvand A. The role of AMPK-dependent pathways in cellular and molecular mechanisms of metformin: a new perspective for treatment and prevention of diseases. Inflammopharmacology 2022; 30:775-788. [PMID: 35419709 PMCID: PMC9007580 DOI: 10.1007/s10787-022-00980-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/20/2022] [Indexed: 02/07/2023]
Abstract
Metformin can suppress gluconeogenesis and reduce blood sugar by activating adenosine monophosphate-activated protein kinase (AMPK) and inducing small heterodimer partner (SHP) expression in the liver cells. The main mechanism of metformin's action is related to its activation of the AMPK enzyme and regulation of the energy balance. AMPK is a heterothermic serine/threonine kinase made of a catalytic alpha subunit and two subunits of beta and a gamma regulator. This enzyme can measure the intracellular ratio of AMP/ATP. If this ratio is high, the amino acid threonine 172 available in its alpha chain would be activated by the phosphorylated liver kinase B1 (LKB1), leading to AMPK activation. Several studies have indicated that apart from its significant role in the reduction of blood glucose level, metformin activates the AMPK enzyme that in turn has various efficient impacts on the regulation of various processes, including controlling inflammatory conditions, altering the differentiation pathway of immune and non-immune cell pathways, and the amelioration of various cancers, liver diseases, inflammatory bowel disease (IBD), kidney diseases, neurological disorders, etc. Metformin's activation of AMPK enables it to control inflammatory conditions, improve oxidative status, regulate the differentiation pathways of various cells, change the pathological process in various diseases, and finally have positive therapeutic effects on them. Due to the activation of AMPK and its role in regulating several subcellular signalling pathways, metformin can be effective in altering the cells' proliferation and differentiation pathways and eventually in the prevention and treatment of certain diseases.
Collapse
Affiliation(s)
- Amin Hasanvand
- Department of Physiology and Pharmacology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
56
|
Wang J, Jiang R, Tan Y, Cheng K. Human pulmonary artery smooth muscle cell dysfunction is regulated by miR-509-5p in hypoxic environment. Cell Cycle 2022; 21:1212-1221. [PMID: 35244512 PMCID: PMC9103279 DOI: 10.1080/15384101.2022.2044147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Reportedly, dysfunction of human pulmonary arterial smooth muscle cells (PASMCs) is associated with the pathogenesis of pulmonary arterial hypertension (PAH). Herein, the role of miR-509-5p in hypoxia-induced PASMCs and the underlying mechanism were explored. PASMCs were cultured under both normoxia and hypoxia conditions. Quantitative real-time polymerase-chain reaction (qPCR) was employed for quantifying the expressions of miR-509-5p and DNMT1 mRNA in the serum of PAH patients and PASMCs. MiR-509-5p mimics and inhibitors were then, respectively, transfected into PAMSCs, and CCK-8 and Transwell assays were utilized to detect PASMCs' proliferation and migration. Flow cytometry was executed for evaluating PASMCs' apoptosis. Interrelation between miR-509-5p and DNMT1 was determined utilizing bioinformatics analysis and dual-luciferase reporter assay. Western blot assay was used to detect the expression of DNMT1 or SOD2. MiR-509-5p in serum samples of patients with PAH as well as hypoxia-induced PASMCs was significantly down-regulated, whereas DNMT1 was markedly up-regulated. MiR-509-5p mimics reduces the proliferation and migration of PASMCs, but promotes the apoptosis; conversely, miR-509-5p inhibitors exerted opposite effects. DNMT1 was identified as a target gene of miR-509-5p, and overexpression of DNMT1 reversed the biological functions of miR-509-5p in regulating the phenotypes of PAMSCs. MiR-509-5p up-regulated the expression of SOD2 by down-regulating DNMT1. MiR-509-5p regulates the proliferation, migration and apoptosis of PASMCs, and restoration of miR-509-5p may be a promising strategy to treat PAH.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Yanlin Tan
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Kuan Cheng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, P.R. China,CONTACT Kuan Cheng Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Xietu Road No.1609, Shanghai200032, P.R.China
| |
Collapse
|
57
|
Zeng Y, Cao J, Li CX, Wang CY, Wu RM, Xu XL. MDM2-Mediated Ubiquitination of RXRβ Contributes to Mitochondrial Damage and Related Inflammation in Atherosclerosis. Int J Mol Sci 2022; 23:ijms23105766. [PMID: 35628577 PMCID: PMC9145909 DOI: 10.3390/ijms23105766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 12/11/2022] Open
Abstract
A novel function of retinoid X receptor beta (RXRβ) in endothelial cells has been reported by us during the formation of atherosclerosis. Here, we extended the study to explore the cellular mechanisms of RXRβ protein stability regulation. In this study, we discovered that murine double minute-2 (MDM2) acts as an E3 ubiquitin ligase to target RXRβ for degradation. The result showed that MDM2 directly interacted with and regulated RXRβ protein stability. MDM2 promoted RXRβ poly-ubiquitination and degradation by proteasomes. Moreover, mutated MDM2 RING domain (C464A) or treatment with an MDM2 inhibitor targeting the RING domain of MDM2 lost the ability of MDM2 to regulate RXRβ protein expression and ubiquitination. Furthermore, treatment with MDM2 inhibitor alleviated oxidized low-density lipoprotein-induced mitochondrial damage, activation of TLR9/NF-κB and NLRP3/caspase-1 pathway and production of pro-inflammatory cytokines in endothelial cells. However, all these beneficial effects were reduced by the transfection of RXRβ siRNA. Moreover, pharmacological inhibition of MDM2 attenuated the development of atherosclerosis and reversed mitochondrial damage and related inflammation in the atherosclerotic process in LDLr-/- mice, along with the increased RXRβ protein expression in the aorta. Therefore, our study uncovers a previously unknown ubiquitination pathway and suggests MDM2-mediated RXRβ ubiquitination as a new therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiao-Le Xu
- Correspondence: ; Tel.: +86-513-8505-1728
| |
Collapse
|
58
|
Liang LJ, Wang D, Yu H, Wang J, Zhang H, Sun BB, Yang FY, Wang Z, Xie DW, Feng RE, Xu KF, Wang GZ, Zhou GB. Transcriptional regulation and small compound targeting of ACE2 in lung epithelial cells. Acta Pharmacol Sin 2022; 43:2895-2904. [PMID: 35468992 PMCID: PMC9035780 DOI: 10.1038/s41401-022-00906-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the receptor of COVID-19 pathogen SARS-CoV-2, but the transcription factors (TFs) that regulate the expression of the gene encoding ACE2 (ACE2) have not been systematically dissected. In this study we evaluated TFs that control ACE2 expression, and screened for small molecule compounds that could modulate ACE2 expression to block SARS-CoV-2 from entry into lung epithelial cells. By searching the online datasets we found that 24 TFs might be ACE2 regulators with signal transducer and activator of transcription 3 (Stat3) as the most significant one. In human normal lung tissues, the expression of ACE2 was positively correlated with phosphorylated Stat3 (p-Stat3). We demonstrated that Stat3 bound ACE2 promoter, and controlled its expression in 16HBE cells stimulated with interleukin 6 (IL-6). To screen for medicinal compounds that could modulate ACE2 expression, we conducted luciferase assay using HLF cells transfected with ACE2 promoter-luciferase constructs. Among the 64 compounds tested, 6-O-angeloylplenolin (6-OAP), a sesquiterpene lactone in Chinese medicinal herb Centipeda minima (CM), represented the most potent ACE2 repressor. 6-OAP (2.5 µM) inhibited the interaction between Stat3 protein and ACE2 promoter, thus suppressed ACE2 transcription. 6-OAP (1.25-5 µM) and its parental medicinal herb CM (0.125%-0.5%) dose-dependently downregulated ACE2 in 16HBE and Beas-2B cells; similar results were observed in the lung tissues of mice following administration of 6-OAP or CM for one month. In addition, 6-OAP/CM dose-dependently reduced IL-6 production and downregulated chemokines including CXCL13 and CX3CL1 in 16HBE cells. Moreover, we found that 6-OAP/CM inhibited the entry of SARS-CoV-2 S protein pseudovirus into target cells. These results suggest that 6-OAP/CM are ACE2 inhibitors that may potentially protect lung epithelial cells from SARS-CoV-2 infection.
Collapse
|
59
|
Skp2 stabilizes Mcl-1 and confers radioresistance in colorectal cancer. Cell Death Dis 2022; 13:249. [PMID: 35301297 PMCID: PMC8930992 DOI: 10.1038/s41419-022-04685-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 11/08/2022]
Abstract
AbstractOverexpression of Skp2 plays a critical role in tumorigenesis and correlates with poor prognosis in human malignancies. Thus, Skp2 has been proposed as an attractive target for anti-tumor interventions. The expression of Skp2 in human colorectal cancer (CRC) and the role of Skp2 in tumorigenic properties and irradiation sensitivities of CRC cells were examined by anchorage-dependent and -independent growth assays, immunoblot, flow cytometry, immunohistochemical staining, ubiquitination analysis, co-immunoprecipitation assay, CRISPR-Cas9-based gene knockout, and xenograft experiments. Skp2 is highly expressed in CRC patient tissues. Blocking Skp2 expression reduces the tumorigenic properties of CRC cells in vitro and in vivo. Depletion of Skp2 confers sensitivity to irradiation of CRC cells. Skp2 deficiency enhances irradiation-induced intrinsic apoptosis by facilitating E3 ligase FBW7-mediated Mcl-1 ubiquitination and degradation. Knockout of Skp2 sensitizes CRC cells to irradiation treatments in vivo. Our findings indicate that Skp2 stabilizes Mcl-1, and targeting Skp2 in combination with traditional radiotherapy might be efficacious in treating CRC.
Collapse
|
60
|
Perez C, Felty Q. Molecular basis of the association between transcription regulators nuclear respiratory factor 1 and inhibitor of DNA binding protein 3 and the development of microvascular lesions. Microvasc Res 2022; 141:104337. [PMID: 35143811 PMCID: PMC8923910 DOI: 10.1016/j.mvr.2022.104337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022]
Abstract
The prognosis of patients with microvascular lesions remains poor because vascular remodeling eventually obliterates the lumen. Here we have focused our efforts on vessel dysfunction in two different organs, the lung and brain. Despite tremendous progress in understanding the importance of blood vessel integrity, gaps remain in our knowledge of the underlying molecular factors contributing to vessel injury, including microvascular lesions. Most of the ongoing research on these lesions have focused on oxidative stress but have not found major molecular targets for the discovery of new treatment or early diagnosis. Herein, we have focused on elucidating the molecular mechanism(s) based on two new emerging molecules NRF1 and ID3, and how they may contribute to microvascular lesions in the lung and brain. Redox sensitive transcriptional activation of target genes depends on not only NRF1, but the recruitment of co-activators such as ID3 to the target gene promoter. Our review highlights the fact that targeting NRF1 and ID3 could be a promising therapeutic approach as they are major players in influencing cell growth, cell repair, senescence, and apoptotic cell death which contribute to vascular lesions. Knowledge about the molecular biology of these processes will be relevant for future therapeutic approaches to not only PAH but cerebral angiopathy and other vascular disorders. Therapies targeting transcription regulators NRF1 or ID3 have the potential for vascular disease-modification because they will address the root causes such as genomic instability and epigenetic changes in vascular lesions. We hope that our findings will serve as a stimulus for further research towards an effective treatment of microvascular lesions.
Collapse
Affiliation(s)
- Christian Perez
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA
| | - Quentin Felty
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.
| |
Collapse
|
61
|
Rossouw TM, Anderson R, Manga P, Feldman C. Emerging Role of Platelet-Endothelium Interactions in the Pathogenesis of Severe SARS-CoV-2 Infection-Associated Myocardial Injury. Front Immunol 2022; 13:776861. [PMID: 35185878 PMCID: PMC8854752 DOI: 10.3389/fimmu.2022.776861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular dysfunction and disease are common and frequently fatal complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Indeed, from early on during the SARS-CoV-2 virus pandemic it was recognized that cardiac complications may occur, even in patients with no underlying cardiac disorders, as part of the acute infection, and that these were associated with more severe disease and increased morbidity and mortality. The most common cardiac complication is acute cardiac injury, defined by significant elevation of cardiac troponins. The potential mechanisms of cardiovascular complications include direct viral myocardial injury, systemic inflammation induced by the virus, sepsis, arrhythmia, myocardial oxygen supply-demand mismatch, electrolyte abnormalities, and hypercoagulability. This review is focused on the prevalence, risk factors and clinical course of COVID-19-related myocardial injury, as well as on current data with regard to disease pathogenesis, specifically the interaction of platelets with the vascular endothelium. The latter section includes consideration of the role of SARS-CoV-2 proteins in triggering development of a generalized endotheliitis that, in turn, drives intense activation of platelets. Most prominently, SARS-CoV-2–induced endotheliitis involves interaction of the viral spike protein with endothelial angiotensin-converting enzyme 2 (ACE2) together with alternative mechanisms that involve the nucleocapsid and viroporin. In addition, the mechanisms by which activated platelets intensify endothelial activation and dysfunction, seemingly driven by release of the platelet-derived calcium-binding proteins, SA100A8 and SA100A9, are described. These events create a SARS-CoV-2–driven cycle of intravascular inflammation and coagulation, which contributes significantly to a poor clinical outcome in patients with severe disease.
Collapse
Affiliation(s)
- Theresa M. Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- *Correspondence: Theresa M. Rossouw,
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Pravin Manga
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
62
|
West J. ACE2 trials suggest remodeling, not dilation, as primary therapeutic effect. Pulm Circ 2022; 12:e12022. [PMID: 35506063 PMCID: PMC9053010 DOI: 10.1002/pul2.12022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- James West
- Vanderbilt University Nashville Tennessee USA
| |
Collapse
|
63
|
Fu Y, Hu L, Ren HW, Zuo Y, Chen S, Zhang QS, Shao C, Ma Y, Wu L, Hao JJ, Wang CZ, Wang Z, Yanagihara R, Deng Y. Prognostic Factors for COVID-19 Hospitalized Patients with Preexisting Type 2 Diabetes. Int J Endocrinol 2022; 2022:9322332. [PMID: 35047039 PMCID: PMC8763039 DOI: 10.1155/2022/9322332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) as a worldwide chronic disease combined with the COVID-19 pandemic prompts the need for improving the management of hospitalized COVID-19 patients with preexisting T2D to reduce complications and the risk of death. This study aimed to identify clinical factors associated with COVID-19 outcomes specifically targeted at T2D patients and build an individualized risk prediction nomogram for risk stratification and early clinical intervention to reduce mortality. METHODS In this retrospective study, the clinical characteristics of 382 confirmed COVID-19 patients, consisting of 108 with and 274 without preexisting T2D, from January 8 to March 7, 2020, in Tianyou Hospital in Wuhan, China, were collected and analyzed. Univariate and multivariate Cox regression models were performed to identify specific clinical factors associated with mortality of COVID-19 patients with T2D. An individualized risk prediction nomogram was developed and evaluated by discrimination and calibration. RESULTS Nearly 15% (16/108) of hospitalized COVID-19 patients with T2D died. Twelve risk factors predictive of mortality were identified. Older age (HR = 1.076, 95% CI = 1.014-1.143, p=0.016), elevated glucose level (HR = 1.153, 95% CI = 1.038-1.28, p=0.0079), increased serum amyloid A (SAA) (HR = 1.007, 95% CI = 1.001-1.014, p=0.022), diabetes treatment with only oral diabetes medication (HR = 0.152, 95%CI = 0.032-0.73, p=0.0036), and oral medication plus insulin (HR = 0.095, 95%CI = 0.019-0.462, p=0.019) were independent prognostic factors. A nomogram based on these prognostic factors was built for early prediction of 7-day, 14-day, and 21-day survival of diabetes patients. High concordance index (C-index) was achieved, and the calibration curves showed the model had good prediction ability within three weeks of COVID-19 onset. CONCLUSIONS By incorporating specific prognostic factors, this study provided a user-friendly graphical risk prediction tool for clinicians to quickly identify high-risk T2D patients hospitalized for COVID-19.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Ling Hu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Hong-Wei Ren
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zuo
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Shaoqiu Chen
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Qiu-Shi Zhang
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Chen Shao
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yao Ma
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Lin Wu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jun-Jie Hao
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Chuan-Zhen Wang
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhanwei Wang
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Richard Yanagihara
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
64
|
Role of Human Antigen R (HuR) in the Regulation of Pulmonary ACE2 Expression. Cells 2021; 11:cells11010022. [PMID: 35011584 PMCID: PMC8750694 DOI: 10.3390/cells11010022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Patients with COPD may be at an increased risk for severe illness from COVID-19 because of ACE2 upregulation, the entry receptor for SARS-CoV-2. Chronic exposure to cigarette smoke, the main risk factor for COPD, increases pulmonary ACE2. How ACE2 expression is controlled is not known but may involve HuR, an RNA binding protein that increases protein expression by stabilizing mRNA. We hypothesized that HuR would increase ACE2 protein expression. We analyzed scRNA-seq data to profile ELAVL1 expression in distinct respiratory cell populations in COVID-19 and COPD patients. HuR expression and cellular localization was evaluated in COPD lung tissue by multiplex immunohistochemistry and in human lung cells by imaging flow cytometry. The regulation of ACE2 expression was evaluated using siRNA-mediated knockdown of HuR. There is a significant positive correlation between ELAVL1 and ACE2 in COPD cells. HuR cytoplasmic localization is higher in smoker and COPD lung tissue; there were also higher levels of cleaved HuR (CP-1). HuR binds to ACE2 mRNA but knockdown of HuR does not change ACE2 protein levels in primary human lung fibroblasts (HLFs). Our work is the first to investigate the association between ACE2 and HuR. Further investigation is needed to understand the mechanistic underpinning behind the regulation of ACE2 expression.
Collapse
|
65
|
Jain PP, Lai N, Xiong M, Chen J, Babicheva A, Zhao T, Parmisano S, Zhao M, Paquin C, Matti M, Powers R, Balistrieri A, Kim NH, Valdez-Jasso D, Thistlethwaite PA, Shyy JYJ, Wang J, Garcia JGN, Makino A, Yuan JXJ. TRPC6, a therapeutic target for pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1161-L1182. [PMID: 34704831 PMCID: PMC8715021 DOI: 10.1152/ajplung.00159.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (PAH) is a fatal and progressive disease. Sustained vasoconstriction due to pulmonary arterial smooth muscle cell (PASMC) contraction and concentric arterial remodeling due partially to PASMC proliferation are the major causes for increased pulmonary vascular resistance and increased pulmonary arterial pressure in patients with precapillary pulmonary hypertension (PH) including PAH and PH due to respiratory diseases or hypoxemia. We and others observed upregulation of TRPC6 channels in PASMCs from patients with PAH. A rise in cytosolic Ca2+ concentration ([Ca2+]cyt) in PASMC triggers PASMC contraction and vasoconstriction, while Ca2+-dependent activation of PI3K/AKT/mTOR pathway is a pivotal signaling cascade for cell proliferation and gene expression. Despite evidence supporting a pathological role of TRPC6, no selective and orally bioavailable TRPC6 antagonist has yet been developed and tested for treatment of PAH or PH. In this study, we sought to investigate whether block of receptor-operated Ca2+ channels using a nonselective blocker of cation channels, 2-aminoethyl diphenylborinate (2-APB, administered intraperitoneally) and a selective blocker of TRPC6, BI-749327 (administered orally) can reverse established PH in mice. The results from the study show that intrapulmonary application of 2-APB (40 µM) or BI-749327 (3-10 µM) significantly and reversibly inhibited acute alveolar hypoxia-induced pulmonary vasoconstriction. Intraperitoneal injection of 2-APB (1 mg/kg per day) significantly attenuated the development of PH and partially reversed established PH in mice. Oral gavage of BI-749327 (30 mg/kg, every day, for 2 wk) reversed established PH by ∼50% via regression of pulmonary vascular remodeling. Furthermore, 2-APB and BI-749327 both significantly inhibited PDGF- and serum-mediated phosphorylation of AKT and mTOR in PASMC. In summary, the receptor-operated and mechanosensitive TRPC6 channel is a good target for developing novel treatment for PAH/PH. BI-749327, a selective TRPC6 blocker, is potentially a novel and effective drug for treating PAH and PH due to respiratory diseases or hypoxemia.
Collapse
MESH Headings
- Animals
- Boron Compounds/pharmacology
- Calcium Signaling
- Cells, Cultured
- Gene Expression Regulation/drug effects
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- TRPC6 Cation Channel/antagonists & inhibitors
- TRPC6 Cation Channel/genetics
- TRPC6 Cation Channel/metabolism
- Vasoconstriction
Collapse
Affiliation(s)
- Pritesh P Jain
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Ning Lai
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingmei Xiong
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Tengteng Zhao
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Sophia Parmisano
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Manjia Zhao
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Cole Paquin
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Moreen Matti
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Ryan Powers
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Angela Balistrieri
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Nick H Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, University of California, San Diego, La Jolla, California
| | - Jian Wang
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
66
|
Rex DAB, Dagamajalu S, Kandasamy RK, Raju R, Prasad TSK. SARS-CoV-2 signaling pathway map: A functional landscape of molecular mechanisms in COVID-19. J Cell Commun Signal 2021; 15:601-608. [PMID: 34181169 PMCID: PMC8237035 DOI: 10.1007/s12079-021-00632-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 has been declared a pandemic by WHO. The clinical manifestation and disease progression in COVID-19 patients varies from minimal symptoms to severe respiratory issues with multiple organ failure. Understanding the mechanism of SARS-CoV-2 interaction with host cells will provide key insights into the effective molecular targets for the development of novel therapeutics. Recent studies have identified virus-mediated phosphorylation or activation of some major signaling pathways, such as ERK1/2, JNK, p38, PI3K/AKT and NF-κB signaling, that potentially elicit the cytokine storm that serves as a major cause of tissue injuries. Several studies highlight the aggressive inflammatory response particularly 'cytokine storm' in SARS-CoV-2 patients. A depiction of host molecular dynamics triggered by SARS-CoV-2 in the form of a network of signaling molecules will be helpful for COVID-19 research. Therefore, we developed the signaling pathway map of SARS-CoV-2 infection using data mined from the recently published literature. This integrated signaling pathway map of SARS-CoV-2 consists of 326 proteins and 73 reactions. These include information pertaining to 1,629 molecular association events, 30 enzyme catalysis events, 43 activation/inhibition events, and 8,531 gene regulation events. The pathway map is publicly available through WikiPathways: https://www.wikipathways.org/index.php/Pathway:WP5115 .
Collapse
Affiliation(s)
- D. A. B. Rex
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| |
Collapse
|
67
|
Rajtik T, Galis P, Bartosova L, Paulis L, Goncalvesova E, Klimas J. Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19. Int J Mol Sci 2021; 22:ijms222312800. [PMID: 34884604 PMCID: PMC8657827 DOI: 10.3390/ijms222312800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Alternative branches of the classical renin–angiotensin–aldosterone system (RAS) represent an important cascade in which angiotensin 2 (AngII) undergoes cleavage via the action of the angiotensin-converting enzyme 2 (ACE2) with subsequent production of Ang(1-7) and other related metabolites eliciting its effects via Mas receptor activation. Generally, this branch of the RAS system is described as its non-canonical alternative arm with counterbalancing actions to the classical RAS, conveying vasodilation, anti-inflammatory, anti-remodeling and anti-proliferative effects. The implication of this branch was proposed for many different diseases, ranging from acute cardiovascular conditions, through chronic respiratory diseases to cancer, nonetheless, hypoxia is one of the most prominent common factors discussed in conjugation with the changes in the activity of alternative RAS branches. The aim of this review is to bring complex insights into the mechanisms behind the various forms of hypoxic insults on the activity of alternative RAS branches based on the different duration of stimuli and causes (acute vs. intermittent vs. chronic), localization and tissue (heart vs. vessels vs. lungs) and clinical relevance of studied phenomenon (experimental vs. clinical condition). Moreover, we provide novel insights into the future strategies utilizing the alternative RAS as a diagnostic tool as well as a promising pharmacological target in serious hypoxia-associated cardiovascular and cardiopulmonary diseases.
Collapse
Affiliation(s)
- Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
- Correspondence: ; Tel.: +42-12-501-17-391
| | - Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Ludovit Paulis
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia;
| | - Eva Goncalvesova
- Department of Heart Failure, Clinic of Cardiology, National Institute of Cardiovascular Diseases, 831 01 Bratislava, Slovakia;
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| |
Collapse
|
68
|
Alam MS, Czajkowsky DM. SARS-CoV-2 infection and oxidative stress: Pathophysiological insight into thrombosis and therapeutic opportunities. Cytokine Growth Factor Rev 2021; 63:44-57. [PMID: 34836751 PMCID: PMC8591899 DOI: 10.1016/j.cytogfr.2021.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023]
Abstract
The current coronavirus disease 2019 (COVID-19) pandemic has presented unprecedented challenges to global health. Although the majority of COVID-19 patients exhibit mild-to-no symptoms, many patients develop severe disease and need immediate hospitalization, with most severe infections associated with a dysregulated immune response attributed to a cytokine storm. Epidemiological studies suggest that overall COVID-19 severity and morbidity correlate with underlying comorbidities, including diabetes, obesity, cardiovascular diseases, and immunosuppressive conditions. Patients with such comorbidities exhibit elevated levels of reactive oxygen species (ROS) and oxidative stress caused by an increased accumulation of angiotensin II and by activation of the NADPH oxidase pathway. Moreover, accumulating evidence suggests that oxidative stress coupled with the cytokine storm contribute to COVID-19 pathogenesis and immunopathogenesis by causing endotheliitis and endothelial cell dysfunction and by activating the blood clotting cascade that results in blood coagulation and microvascular thrombosis. In this review, we survey the mechanisms of how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces oxidative stress and the consequences of this stress on patient health. We further shed light on aspects of the host immunity that are crucial to prevent the disease during the early phase of infection. A better understanding of the disease pathophysiology as well as preventive measures aimed at lowering ROS levels may pave the way to mitigate SARS-CoV-2-induced complications and decrease mortality.
Collapse
Affiliation(s)
- Mohammad Shah Alam
- Department of Anatomy and Histology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh.
| | - Daniel M Czajkowsky
- Bio-ID Centre, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
69
|
Wang X, Li Y, He M, Kong X, Jiang P, Liu X, Diao L, Zhang X, Li H, Ling X, Xia S, Liu Z, Liu Y, Cui CP, Wang Y, Tang L, Zhang L, He F, Li D. UbiBrowser 2.0: a comprehensive resource for proteome-wide known and predicted ubiquitin ligase/deubiquitinase-substrate interactions in eukaryotic species. Nucleic Acids Res 2021; 50:D719-D728. [PMID: 34669962 PMCID: PMC8728189 DOI: 10.1093/nar/gkab962] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
As an important post-translational modification, ubiquitination mediates ∼80% of protein degradation in eukaryotes. The degree of protein ubiquitination is tightly determined by the delicate balance between specific ubiquitin ligase (E3)-mediated ubiquitination and deubiquitinase-mediated deubiquitination. In 2017, we developed UbiBrowser 1.0, which is an integrated database for predicted human proteome-wide E3-substrate interactions. Here, to meet the urgent requirement of proteome-wide E3/deubiquitinase-substrate interactions (ESIs/DSIs) in multiple organisms, we updated UbiBrowser to version 2.0 (http://ubibrowser.ncpsb.org.cn). Using an improved protocol, we collected 4068/967 known ESIs/DSIs by manual curation, and we predicted about 2.2 million highly confident ESIs/DSIs in 39 organisms, with >210-fold increase in total data volume. In addition, we made several new features in the updated version: (i) it allows exploring proteins' upstream E3 ligases and deubiquitinases simultaneously; (ii) it has significantly increased species coverage; (iii) it presents a uniform confidence scoring system to rank predicted ESIs/DSIs. To facilitate the usage of UbiBrowser 2.0, we also redesigned the web interface for exploring these known and predicted ESIs/DSIs, and added functions of 'Browse', 'Download' and 'Application Programming Interface'. We believe that UbiBrowser 2.0, as a discovery tool, will contribute to the study of protein ubiquitination and the development of drug targets for complex diseases.
Collapse
Affiliation(s)
- Xun Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Mengqi He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiangren Kong
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Peng Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xi Liu
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Lihong Diao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xinlei Zhang
- Beijing Geneworks Technology Co.,Ltd., Beijing 100101, China
| | - Honglei Li
- Beijing Geneworks Technology Co.,Ltd., Beijing 100101, China
| | - Xinping Ling
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Simin Xia
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongyang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yuan Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Liujun Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,College of Life Sciences, Hebei University, Baoding 071002, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
70
|
Tian Q, Fan X, Ma J, Li D, Han Y, Yin X, Wang H, Huang T, Wang Z, Shentu Y, Xue F, Du C, Wang Y, Mao S, Fan J, Gong Y. Critical role of VGLL4 in the regulation of chronic normobaric hypoxia-induced pulmonary hypertension in mice. FASEB J 2021; 35:e21822. [PMID: 34314061 DOI: 10.1096/fj.202002650rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Pulmonary hypertension (PH), a rare but deadly cardiopulmonary disorder, is characterized by extensive remodeling of pulmonary arteries resulting from enhancement of pulmonary artery smooth muscle cell proliferation and suppressed apoptosis; however, the underlying pathophysiological mechanisms remain largely unknown. Recently, epigenetics has gained increasing prominence in the development of PH. We aimed to investigate the role of vestigial-like family member 4 (VGLL4) in chronic normobaric hypoxia (CNH)-induced PH and to address whether it is associated with epigenetic regulation. The rodent model of PH was established by CNH treatment (10% O2 , 23 hours/day). Western blot, quantitative reverse transcription polymerase chain reaction, immunofluorescence, immunoprecipitation, and adeno-associated virus tests were performed to explore the potential mechanisms involved in CNH-induced PH in mice. VGLL4 expression was upregulated and correlated with CNH in PH mouse lung tissues in a time-dependent manner. VGLL4 colocalized with α-smooth muscle actin in cultured pulmonary arterial smooth muscle cells (PASMCs), and VGLL4 immunoactivity was increased in PASMCs following hypoxia exposure in vitro. VGLL4 knockdown attenuated CNH-induced PH and pulmonary artery remodeling by blunting signal transducer and activator of transcription 3 (STAT3) signaling; conversely, VGLL4 overexpression exacerbated the development of PH. CNH enhanced the acetylation of VGLL4 and increased the interaction of ac-H3K9/VGLL4 and ac-H3K9/STAT3 in the lung tissues, and levels of ac-H3K9, p-STAT3/STAT3, and proliferation-associated protein levels were markedly up-regulated, whereas apoptosis-related protein levels were significantly downregulated, in the lung tissues of mice with CNH-induced PH. Notably, abrogation of VGLL4 acetylation reversed CNH-induced PH and pulmonary artery remodeling and suppressed STAT3 signaling. Finally, STAT3 knockdown alleviated CNH-induced PH. In conclusion, VGLL4 acetylation upregulation could contribute to CNH-induced PH and pulmonary artery remodeling via STAT3 signaling, and abrogation of VGLL4 acetylation reversed CNH-induced PH. Pharmacological or genetic deletion of VGLL4 might be a potential target for therapeutic interventions in CNH-induced PH.
Collapse
Affiliation(s)
- Qiuyun Tian
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianshe Ma
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dantong Li
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yujiao Han
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xianghong Yin
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Tingting Huang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhenglu Wang
- Renji College, Wenzhou Medical University, Wenzhou, China
| | - Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Xue
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongyu Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunzhong Mao
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
71
|
Tovo PA, Garazzino S, Daprà V, Pruccoli G, Calvi C, Mignone F, Alliaudi C, Denina M, Scolfaro C, Zoppo M, Licciardi F, Ramenghi U, Galliano I, Bergallo M. COVID-19 in Children: Expressions of Type I/II/III Interferons, TRIM28, SETDB1, and Endogenous Retroviruses in Mild and Severe Cases. Int J Mol Sci 2021; 22:7481. [PMID: 34299101 PMCID: PMC8303145 DOI: 10.3390/ijms22147481] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023] Open
Abstract
Children with the new coronavirus disease 2019 (COVID-19) have milder symptoms and a better prognosis than adult patients. Several investigations assessed type I, II, and III interferon (IFN) signatures in SARS-CoV-2 infected adults, however no data are available for pediatric patients. TRIM28 and SETDB1 regulate the transcription of multiple genes involved in the immune response as well as of human endogenous retroviruses (HERVs). Exogenous viral infections can trigger the activation of HERVs, which in turn can induce inflammatory and immune reactions. Despite the potential cross-talks between SARS-CoV-2 infection and TRIM28, SETDB1, and HERVs, information on their expressions in COVID-19 patients is lacking. We assessed, through a PCR real time Taqman amplification assay, the transcription levels of six IFN-I stimulated genes, IFN-II and three of its sensitive genes, three IFN-lIIs, as well as of TRIM28, SETDB1, pol genes of HERV-H, -K, and -W families, and of env genes of Syncytin (SYN)1, SYN2, and multiple sclerosis-associated retrovirus (MRSV) in peripheral blood from COVID-19 children and in control uninfected subjects. Higher expression levels of IFN-I and IFN-II inducible genes were observed in 36 COVID-19 children with mild or moderate disease as compared to uninfected controls, whereas their concentrations decreased in 17 children with severe disease and in 11 with multisystem inflammatory syndrome (MIS-C). Similar findings were found for the expression of TRIM-28, SETDB1, and every HERV gene. Positive correlations emerged between the transcriptional levels of type I and II IFNs, TRIM28, SETDB1, and HERVs in COVID-19 patients. IFN-III expressions were comparable in each group of subjects. This preserved induction of IFN-λs could contribute to the better control of the infection in children as compared to adults, in whom IFN-III deficiency has been reported. The upregulation of IFN-I, IFN-II, TRIM28, SETDB1, and HERVs in children with mild symptoms, their declines in severe cases or with MIS-C, and the positive correlations of their transcription in SARS-CoV-2-infected children suggest that they may play important roles in conditioning the evolution of the infection.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
| | - Silvia Garazzino
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (S.G.); (F.M.); (C.S.); (M.Z.)
| | - Valentina Daprà
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, 10126 Turin, Italy;
| | - Giulia Pruccoli
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
| | - Cristina Calvi
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, 10126 Turin, Italy;
| | - Federica Mignone
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (S.G.); (F.M.); (C.S.); (M.Z.)
| | - Carla Alliaudi
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, 10126 Turin, Italy;
| | - Marco Denina
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
| | - Carlo Scolfaro
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (S.G.); (F.M.); (C.S.); (M.Z.)
| | - Marisa Zoppo
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy; (S.G.); (F.M.); (C.S.); (M.Z.)
| | - Francesco Licciardi
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
| | - Ugo Ramenghi
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
| | - Ilaria Galliano
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, 10126 Turin, Italy;
| | - Massimiliano Bergallo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (G.P.); (C.C.); (C.A.); (M.D.); (F.L.); (U.R.); (I.G.)
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, 10126 Turin, Italy;
| |
Collapse
|
72
|
Wang Y, Han D, Zhou T, Chen C, Cao H, Zhang JZ, Ma N, Liu C, Song M, Shi J, Jin X, Cao F, Dong N. DUSP26 induces aortic valve calcification by antagonizing MDM2-mediated ubiquitination of DPP4 in human valvular interstitial cells. Eur Heart J 2021; 42:2935-2951. [PMID: 34179958 DOI: 10.1093/eurheartj/ehab316] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/21/2021] [Accepted: 05/07/2021] [Indexed: 12/21/2022] Open
Abstract
AIMS The morbidity and mortality rates of calcific aortic valve disease (CAVD) remain high while treatment options are limited. Here, we evaluated the role and therapeutic value of dual-specificity phosphatase 26 (DUSP26) in CAVD. METHODS AND RESULTS Microarray profiling of human calcific aortic valves and normal controls demonstrated that DUSP26 was significantly up-regulated in calcific aortic valves. ApoE-/- mice fed a normal diet or a high cholesterol diet (HCD) were infected with adeno-associated virus serotype 2 carrying DUSP26 short-hairpin RNA to examine the effects of DUSP26 silencing on aortic valve calcification. DUSP26 silencing ameliorated aortic valve calcification in HCD-treated ApoE-/- mice, as evidenced by reduced thickness and calcium deposition in the aortic valve leaflets, improved echocardiographic parameters (decreased peak transvalvular jet velocity and mean transvalvular pressure gradient, as well as increased aortic valve area), and decreased levels of osteogenic markers (Runx2, osterix, and osteocalcin) in the aortic valves. These results were confirmed in osteogenic medium-induced human valvular interstitial cells. Immunoprecipitation, liquid chromatography-tandem mass spectrometry, and functional assays revealed that dipeptidyl peptidase-4 (DPP4) interacted with DUSP26 to mediate the procalcific effects of DUSP26. High N6-methyladenosine levels up-regulated DUSP26 in CAVD; in turn, DUSP26 activated DPP4 by antagonizing mouse double minute 2-mediated ubiquitination and degradation of DPP4, thereby promoting CAVD progression. CONCLUSION DUSP26 promotes aortic valve calcification by inhibiting DPP4 degradation. Our findings identify a previously unrecognized mechanism of DPP4 up-regulation in CAVD, suggesting that DUSP26 silencing or inhibition is a viable therapeutic strategy to impede CAVD progression.
Collapse
Affiliation(s)
- Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Dong Han
- National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, 28# Fuxing Road, Beijing 100853, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Cheng Chen
- Institute of Geriatrics, National Clinical Research Center for Geriatrics Disease, Chinese PLA General Hospital, 28# Fuxing Road, Beijing 100853, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Joe Z Zhang
- Stanford Cardiovascular Institute, Stanford School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Ning Ma
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), 96# Xingdao South Road, Haizhu District, Guangzhou, Guangdong 510320, China
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Moshi Song
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, 1# Beichen West Road, Beijing 100101, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, 139# Renmin middle road, Changsha, Hunan 410011, China
| | - Feng Cao
- National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, 28# Fuxing Road, Beijing 100853, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei 430022, China
| |
Collapse
|
73
|
Gonzalez SM, Siddik AB, Su RC. Regulated Intramembrane Proteolysis of ACE2: A Potential Mechanism Contributing to COVID-19 Pathogenesis? Front Immunol 2021; 12:612807. [PMID: 34163462 PMCID: PMC8215698 DOI: 10.3389/fimmu.2021.612807] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/07/2021] [Indexed: 12/22/2022] Open
Abstract
Since being identified as a key receptor for SARS-CoV-2, Angiotensin converting enzyme 2 (ACE2) has been studied as one of the potential targets for the development of preventative and/or treatment options. Tissue expression of ACE2 and the amino acids interacting with the spike protein of SARS-CoV-2 have been mapped. Furthermore, the recombinant soluble extracellular domain of ACE2 is already in phase 2 trials as a treatment for SARS-CoV-2 infection. Most studies have continued to focus on the ACE2 extracellular domain, which is known to play key roles in the renin angiotensin system and in amino acid uptake. However, few also found ACE2 to have an immune-modulatory function and its intracellular tail may be one of the signaling molecules in regulating cellular activation. The implication of its immune-modulatory role in preventing the cytokine-storm, observed in severe COVID-19 disease outcomes requires further investigation. This review focuses on the regulated proteolytic cleavage of ACE2 upon binding to inducer(s), such as the spike protein of SARS-CoV, the potential of cleaved ACE2 intracellular subdomain in regulating cellular function, and the ACE2's immune-modulatory function. This knowledge is critical for targeting ACE2 levels for developing prophylactic treatment or preventative measures in SARS-CoV infections.
Collapse
Affiliation(s)
- Sandra M. Gonzalez
- Department of Medical Microbiology and Infectious Diseases, University of Manitobag, Winnipe, MB, Canada
| | - Abu Bakar Siddik
- Department of Medical Microbiology and Infectious Diseases, University of Manitobag, Winnipe, MB, Canada
- National HIV and Retrovirology Laboratories, J.C. Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Ruey-Chyi Su
- Department of Medical Microbiology and Infectious Diseases, University of Manitobag, Winnipe, MB, Canada
- National HIV and Retrovirology Laboratories, J.C. Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada
| |
Collapse
|
74
|
Fang S, Cheng Y, Deng F, Zhang B. RNF34 ablation promotes cerebrovascular remodeling and hypertension by increasing NADPH-derived ROS generation. Neurobiol Dis 2021; 156:105396. [PMID: 34015492 DOI: 10.1016/j.nbd.2021.105396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 10/21/2022] Open
Abstract
Cerebrovascular remodeling is the most common cause of hypertension and stroke. Ubiquitin E3 ligase RING finger protein 34 (RNF34) is suggested to be associated with the development of multiple neurological diseases. However, the importance of RNF34 in cerebrovascular remodeling and hypertension is poorly understood. Herein, we used mice with a global RNF34 knockout as well as RNF34 floxed mice to delete RNF34 in endothelial cells and smooth muscle cells (SMCs). Our results showed that global RNF34 knockout mice substantially promoted angiotensin II (AngII)-induced middle cerebral artery (MCA) remodeling, hypertension, and neurological dysfunction. Endothelial cell RNF34 did not regulate the development of hypertension. Rather, SMC RNF34 expression is a critical regulator of hypertension and MCA remodeling. Loss of RNF34 enhanced AngII-induced mouse brain vascular SMCs (MBVSMCs) proliferation, migration and invasion. Furthermore, MCA and MBVSMCs from SMC RNF34-deficient mice showed increased superoxide anion and reactive oxygen species (ROS) generation as well as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, but exhibited no marked effect on mitochondria-derived ROS. Knockout of RNF34 promoted p22phox expression, leading to increased binding of p22phox/p47phox and p22phox/NOX2, and eventually NADPH oxidase complex formation. Immunoprecipitation assay identified that RNF34 interacted with p22phox. RNF34 deletion increased p22phox protein stability by inhibiting ubiquitin-mediated degradation. Blockade of NADPH oxidase activity or knockdown of p22phox significantly abolished the effects of RNF34 deletion on cerebrovascular remodeling and hypertension. Collectively, our study demonstrates that SMC RNF34 deficiency promotes cerebrovascular SMC hyperplasia and remodeling by increased NADPH-derived ROS generation via reducing p22phox ubiquitin-dependent degradation.
Collapse
Affiliation(s)
- Shaokuan Fang
- Department of Neurology, The First Teaching Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Yingying Cheng
- Department of Neurology, The First Teaching Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Fang Deng
- Department of Neurology, The First Teaching Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Beilin Zhang
- Department of Neurology, The First Teaching Hospital of Jilin University, Changchun 130021, Jilin, China.
| |
Collapse
|
75
|
Kostyunina DS, McLoughlin P. Sex Dimorphism in Pulmonary Hypertension: The Role of the Sex Chromosomes. Antioxidants (Basel) 2021; 10:779. [PMID: 34068984 PMCID: PMC8156365 DOI: 10.3390/antiox10050779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023] Open
Abstract
Pulmonary hypertension (PH) is a condition characterised by an abnormal elevation of pulmonary artery pressure caused by an increased pulmonary vascular resistance, frequently leading to right ventricular failure and reduced survival. Marked sexual dimorphism is observed in patients with pulmonary arterial hypertension, a form of pulmonary hypertension with a particularly severe clinical course. The incidence in females is 2-4 times greater than in males, although the disease is less severe in females. We review the contribution of the sex chromosomes to this sex dimorphism highlighting the impact of proteins, microRNAs and long non-coding RNAs encoded on the X and Y chromosomes. These genes are centrally involved in the cellular pathways that cause increased pulmonary vascular resistance including the production of reactive oxygen species, altered metabolism, apoptosis, inflammation, vasoconstriction and vascular remodelling. The interaction with genetic mutations on autosomal genes that cause heritable pulmonary arterial hypertension such as bone morphogenetic protein 2 (BMPR2) are examined. The mechanisms that can lead to differences in the expression of genes located on the X chromosomes between females and males are also reviewed. A better understanding of the mechanisms of sex dimorphism in this disease will contribute to the development of more effective therapies for both women and men.
Collapse
Affiliation(s)
| | - Paul McLoughlin
- Conway Institute, School of Medicine, University College Dublin, Dublin D04 V1W8, Ireland;
| |
Collapse
|
76
|
Su S, Chen J, Wang Y, Wong LM, Zhu Z, Jiang G, Liu P. Lenalidomide downregulates ACE2 protein abundance to alleviate infection by SARS-CoV-2 spike protein conditioned pseudoviruses. Signal Transduct Target Ther 2021; 6:182. [PMID: 33972500 PMCID: PMC8107201 DOI: 10.1038/s41392-021-00608-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/28/2022] Open
Affiliation(s)
- Siyuan Su
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianfeng Chen
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ying Wang
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lilly M Wong
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhichuan Zhu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Guochun Jiang
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
77
|
Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev 2021; 35:575-601. [PMID: 33888565 PMCID: PMC8091979 DOI: 10.1101/gad.347872.120] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, Klein et al. discuss the p53-independent roles of MDM2 and MDMX. First, they review the structural and functional features of MDM2 and MDMX proteins separately and together that could be relevant to their p53-independent activities. Following this, they summarize how these two proteins are regulated and how they can function in cells that lack p53. Most well studied as proteins that restrain the p53 tumor suppressor protein, MDM2 and MDMX have rich lives outside of their relationship to p53. There is much to learn about how these two proteins are regulated and how they can function in cells that lack p53. Regulation of MDM2 and MDMX, which takes place at the level of transcription, post-transcription, and protein modification, can be very intricate and is context-dependent. Equally complex are the myriad roles that these two proteins play in cells that lack wild-type p53; while many of these independent outcomes are consistent with oncogenic transformation, in some settings their functions could also be tumor suppressive. Since numerous small molecules that affect MDM2 and MDMX have been developed for therapeutic outcomes, most if not all designed to prevent their restraint of p53, it will be essential to understand how these diverse molecules might affect the p53-independent activities of MDM2 and MDMX.
Collapse
Affiliation(s)
- Alyssa M Klein
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, New York 10032, USA
| | | | - Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
78
|
Lei Y, Zhang J, Schiavon CR, He M, Chen L, Shen H, Zhang Y, Yin Q, Cho Y, Andrade L, Shadel GS, Hepokoski M, Lei T, Wang H, Zhang J, Yuan JXJ, Malhotra A, Manor U, Wang S, Yuan ZY, Shyy JYJ. SARS-CoV-2 Spike Protein Impairs Endothelial Function via Downregulation of ACE 2. Circ Res 2021; 128:1323-1326. [PMID: 33784827 PMCID: PMC8091897 DOI: 10.1161/circresaha.121.318902] [Citation(s) in RCA: 292] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yuyang Lei
- Cardiology, First Affiliated Hospital of Xi'an Jiaotong University (Y.L., Jiao Zhang, Z.-Y.Y.).,Cardiovascular Research Center, School of Basic Medical Sciences (Y.L., Jiao Zhang, L.C., Q.Y., S.W.)
| | - Jiao Zhang
- Cardiology, First Affiliated Hospital of Xi'an Jiaotong University (Y.L., Jiao Zhang, Z.-Y.Y.).,Cardiovascular Research Center, School of Basic Medical Sciences (Y.L., Jiao Zhang, L.C., Q.Y., S.W.).,Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA
| | - Cara R Schiavon
- Waitt Advanced Biophotonics Center (C.R.S., L.A., U.M.).,Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA (C.R.S., G.S.S.)
| | - Ming He
- Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA
| | - Lili Chen
- Cardiovascular Research Center, School of Basic Medical Sciences (Y.L., Jiao Zhang, L.C., Q.Y., S.W.)
| | - Hui Shen
- Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA.,Cardiology, the Affiliated Hospital of Yangzhou University (H.S.)
| | - Yichi Zhang
- Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA
| | - Qian Yin
- Cardiovascular Research Center, School of Basic Medical Sciences (Y.L., Jiao Zhang, L.C., Q.Y., S.W.)
| | - Yoshitake Cho
- Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA
| | | | - Gerald S Shadel
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA (C.R.S., G.S.S.)
| | - Mark Hepokoski
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine (M. Hepokoski, J.X.-J.Y., A.M.), University of California, San Diego, La Jolla, CA
| | - Ting Lei
- Pathology, School of Basic Medical Sciences (T.L.), University of California, San Diego, La Jolla, CA
| | - Hongliang Wang
- Pathogen Biology and Immunology, School of Basic Medical Sciences (H.W.), University of California, San Diego, La Jolla, CA
| | - Jin Zhang
- Pharmacology (Jin Zhang), University of California, San Diego, La Jolla, CA
| | - Jason X-J Yuan
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine (M. Hepokoski, J.X.-J.Y., A.M.), University of California, San Diego, La Jolla, CA
| | - Atul Malhotra
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine (M. Hepokoski, J.X.-J.Y., A.M.), University of California, San Diego, La Jolla, CA
| | - Uri Manor
- Waitt Advanced Biophotonics Center (C.R.S., L.A., U.M.)
| | - Shengpeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences (Y.L., Jiao Zhang, L.C., Q.Y., S.W.)
| | - Zu-Yi Yuan
- Cardiology, First Affiliated Hospital of Xi'an Jiaotong University (Y.L., Jiao Zhang, Z.-Y.Y.)
| | - John Y-J Shyy
- Xi'an Jiaotong University Health Science Center. Cardiology, Department of Medicine (Jiao Zhang, M. He, H.S., Y.Z., Y.C., J.Y.-J.S.), University of California, San Diego, La Jolla, CA
| |
Collapse
|
79
|
Mohamed T, Abdul-Hafez A, Uhal BD. Regulation of ACE-2 enzyme by hyperoxia in lung epithelial cells by post-translational modification. JOURNAL OF LUNG, PULMONARY & RESPIRATORY RESEARCH 2021; 8:47-52. [PMID: 34825051 PMCID: PMC8612072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Bronchopulmonary Dysplasia (BPD) occurs in premature neonates with respiratory distress who require supplemental oxygen in the first days after birth. BPD involves uniform arrest of alveolar development and variable interstitial cellularity and/or fibroproliferation. Previous studies by our lab showed that the enzyme, angiotensin converting enzyme-2 (ACE-2) and its product Ang1-7 exerting action on the receptor Mas oncogene in what is known as ACE-2/Mas axis is protective to lung cells. We also showed that ACE-2 is expressed in fetal human lung fibroblasts but is significantly decreased by hyperoxic gas lung injury, an effect caused by ACE-2 enzyme shedding mediated by TNF-alpha-converting enzyme (TACE/ADAM17). However, no reports yet exist about the regulation of ACE-2 in the alveolar epithelia in hyperoxic lung injury. OBJECTIVE In this study we aim to define the effects of hyperoxic lung injury on the protective ACE-2 enzyme in the human lung alveolar epithelial cell line A549. DESIGN/METHODS Cultured A549 cells were exposed to hyperoxia (95% O2) or normoxia (21% O2) for 3 or 7 days in serum-free nutrient media. Cells were lysed and culture media were collected to test for cellular ACE-2 enzymatic activity and for ACE-2, Mas receptor, TACE/ADAM17, and ubiquitin proteins abundance by immunoblotting. Cells were harvested in Trizol for RNA extraction and ACE-2 qRT-PCR. Whole cell extracts of A549 cell line was used for ACE-2 immunoprecipitation and subsequent ubiquitin immunoblotting. RESULTS Total ubiquitinated proteins were increased by hyperoxia treatment, while ACE-2 and Mas receptor proteins abundance and ACE-2 enzymatic activity were decreased significantly in A549 cells exposed to hyperoxia relative to the normoxia controls. The percent decrease in ACE-2 activity corresponded with increased time of hyperoxic gas exposure. However, in contrast to our data from lung fibroblasts, no significant change was noted in ACE-2 protein released into the media or in ACE-2 mRNA levels by the hyperoxic treatment. Ubiquitin immunoreactive bands were detectable in the ACE-2 immunoprecipitate. CONCLUSIONS These data suggest that hyperoxic exposure of the lung epithelial cells decreases the protective enzyme ACE-2 by cell type specific mechanisms independent of shedding by TACE/ADAM17. The data also suggest a regulatory level of ACE-2 downstream of transcription may involve ACE-2 ubiquitination and targeting for degradation.
Collapse
Affiliation(s)
- Tarek Mohamed
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Amal Abdul-Hafez
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, USA
| |
Collapse
|
80
|
SARS-CoV-2 Spike Protein Impairs Endothelial Function via Downregulation of ACE2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33300001 DOI: 10.1101/2020.12.04.409144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Coronavirus disease 2019 (COVID-19) includes the cardiovascular complications in addition to respiratory disease. SARS-CoV-2 infection impairs endothelial function and induces vascular inflammation, leading to endotheliitis. SARS-CoV-2 infection relies on the binding of Spike glycoprotein (S protein) to angiotensin converting enzyme 2 (ACE2) in the host cells. We show here that S protein alone can damage vascular endothelial cells (ECs) in vitro and in vivo, manifested by impaired mitochondrial function, decreased ACE2 expression and eNOS activity, and increased glycolysis. The underlying mechanism involves S protein downregulation of AMPK and upregulation of MDM2, causing ACE2 destabilization. Thus, the S protein-exerted vascular endothelial damage via ACE2 downregulation overrides the decreased virus infectivity.
Collapse
|