1
|
Mallah N, Visos-Varela I, Takkouche B, Bugarín-González R, Piñeiro-Lamas M, Herdeiro T, Zapata-Cachafeiro M, Rodríguez-Fernández A, Salgado-Barreira A, Figueiras A. The role of traditional NSAIDs and selective COX-2 inhibitors on COVID-19 outcomes: a real-world data study. Inflammopharmacology 2024; 32:3697-3705. [PMID: 39312097 DOI: 10.1007/s10787-024-01568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/31/2024] [Indexed: 11/10/2024]
Abstract
The relation between use of nonsteroidal anti-inflammatory drugs (NSAIDs) and severity of COVID-19 has been the subject to debate since the outbreak of the pandemic. Despite speculations about the possible harmful or protective effects, the position currently most supported by the scientific community is that there is no association between use of NSAIDs and COVID-19 outcomes. With the aim of contributing to increase the body of evidence on this issue, we conducted a case-control study using real-world data to investigate the association between prior use of NSAIDs, by active ingredient and type (traditional NSAIDs and selective COX-2 inhibitors), and important COVID-19-related outcomes, including susceptibility, PCR + patient progression, and hospitalisation. Our findings suggest that, in general, the use of traditional NSAIDs is not associated with any adverse COVID-19 outcome. However, we observed a possible association between diclofenac and a higher risk of PCR + patient progression. Our results also suggest that selective COX-2 inhibitors might be related with a reduction in the risk of PCR + patient progression. These results suggest that, with the possible exception of diclofenac, the use of NSAIDs should not be advised against for relief of symptoms in patients with COVID-19. In addition, they support the importance of continue to investigate the treatment potential of selective COX-2 inhibitors in the management of COVID-19, something that could have significant implications for the treatment of this disease and other viral infections.
Collapse
Affiliation(s)
- Narmeen Mallah
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Institute of Health Research of Santiago de Compostela de Compostela, Santiago de Compostela, Spain
- WHO Collaborating Centre for Vaccine Safety, Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
| | - Irene Visos-Varela
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Institute of Health Research of Santiago de Compostela de Compostela, Santiago de Compostela, Spain.
- WHO Collaborating Centre for Vaccine Safety, Santiago de Compostela, Spain.
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain.
| | - Bahi Takkouche
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Rosendo Bugarín-González
- Centro de Salud de Monforte de Lemos, Área Sanitaria de Lugo, A Mariña e Monforte de Lemos, SERGAS, Monforte de Lemos, Lugo, Spain
| | - María Piñeiro-Lamas
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
| | - Teresa Herdeiro
- Department of Medical Sciences, iBiMED-Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Maruxa Zapata-Cachafeiro
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Almudena Rodríguez-Fernández
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Angel Salgado-Barreira
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Adolfo Figueiras
- Consortium for Biomedical Research in Epidemiology & Public Health (CIBER en Epidemiología y Salud Pública-CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/ San Francisco s/n, 15782, Santiago de Compostela, Spain
- Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
2
|
Cai P, Li W, Xu Y, Wang H. Drp1 and neuroinflammation: Deciphering the interplay between mitochondrial dynamics imbalance and inflammation in neurodegenerative diseases. Neurobiol Dis 2024; 198:106561. [PMID: 38857809 DOI: 10.1016/j.nbd.2024.106561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
Neuroinflammation and mitochondrial dysfunction are closely intertwined with the pathophysiology of neurological disorders. Recent studies have elucidated profound alterations in mitochondrial dynamics across a spectrum of neurological disorders. Dynamin-related protein 1 (DRP1) emerges as a pivotal regulator of mitochondrial fission, with its dysregulation disrupting mitochondrial homeostasis and fueling neuroinflammation, thereby exacerbating disease severity. In addition to its role in mitochondrial dynamics, DRP1 plays a crucial role in modulating inflammation-related pathways. This review synthesizes important functions of DRP1 in the central nervous system (CNS) and the impact of epigenetic modification on the progression of neurodegenerative diseases. The intricate interplay between neuroinflammation and DRP1 in microglia and astrocytes, central contributors to neuroinflammation, is expounded upon. Furthermore, the use of DRP1 inhibitors to influence the activation of microglia and astrocytes, as well as their involvement in processes such as mitophagy, mitochondrial oxidative stress, and calcium ion transport in CNS-mediated neuroinflammation, is scrutinized. The modulation of microglia to astrocyte crosstalk by DRP1 and its role in inflammatory neurodegeneration is also highlighted. Overall, targeting DRP1 presents a promising avenue for ameliorating neuroinflammation and enhancing the therapeutic management of neurological disorders.
Collapse
Affiliation(s)
- Peiyang Cai
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wuhao Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Ye Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China..
| |
Collapse
|
3
|
Elnegris HM, Abdelrahman AA, El-Roghy ES. The potential therapeutic effects of exosomes derived from bone marrow mesenchymal stem cells on ileum injury of a rat sepsis model (histological and immunohistochemical study). Ultrastruct Pathol 2024; 48:274-296. [PMID: 38946300 DOI: 10.1080/01913123.2024.2368011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Sepsis denotes a serious high mortality concern. The study was designed to evaluate the effect of mesenchymal stem cell exosomes (MSC-exosomes) on the evolution of the animal model of sepsis. In this study, 36 rats were distributed into three groups, (I) controls, (II) LPS-treated, and (III) LPS+MSC-EVs. Sepsis was simulated by administering E. coli-LPS to the laboratory animals. Group III was given MSC-exosomes four hours after the LPS injection. Forty-eight hours later rats were sacrificed. Ileum samples were excised, and processed for the histological assessment, immunohistochemical identification of CD44, and inducible nitric oxide synthase (iNOS). Ileum homogenate was used to estimate tumor necrosis factor α (TNF α) besides Cyclooxygenase-2 (COX 2). PCR was used for the detection of interleukin 1α (IL‑1α), and interleukin 17 (IL‑17). Statistical and morphometrical analysis was done. The LPS-treated group showed increased TNF-α, IL‑1α, IL‑17, and decreased COX 2. LPS administration led to cytoplasmic vacuolization of enterocytes, an increase in the vasculature, and cellular infiltrations invaded the lamina propria. There was a significant rise in goblet cells and the proportion of collagen fibers. Ultrastructurally, the enterocytes displayed nuclear irregularity, rough endoplasmic reticulum (rER) dilatation, and increased mitochondria number. Sepsis induces a significant increase in iNOS and a decrease in CD44 immune expressions. LPS+MSC-EVs group restored normal ileum structure and revealed a significant elevation in CD44 and a reduction in iNOS immunoreactions. LPS-sepsis induced an obvious ileum inflammatory deterioration ameliorated by MSC-exosomes, mostly through their antioxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Heba M Elnegris
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Department of Histology and Cell Biology, Faculty of Medicine, Badr University in Cairo, Cairo, Egypt
| | - Abeer A Abdelrahman
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman S El-Roghy
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shebin el Kom, Egypt
| |
Collapse
|
4
|
Liu J, Mai T, Ren H, Chang Y, Li C, Lv G, Zheng D, Liao X, Yu Y, Zhang F, Zhao S, Liu X, Liu S, Zhao H, Luo B, Liu C, Huang E. Arrhythmia onsets triggered by acute myocardial ischemia are not mediated by lysophosphoglycerides accumulation in ventricular myocardium. Sci Rep 2024; 14:9589. [PMID: 38670979 PMCID: PMC11053080 DOI: 10.1038/s41598-024-57047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Lysophosphoglycerides (LPLs) have been reported to accumulate in myocardium and serve as a cause of arrhythmias in acute myocardial ischemia. However, in this study we found that LPLs level in the ventricular myocardium was decreased by the onset of acute myocardial ischemia in vivo in rats. Decreasing of LPLs level in left ventricular myocardium, but not right, was observed within 26 min of left myocardial ischemia, regardless of whether arrhythmias were triggered. Lower LPLs level in the ventricular myocardium was also observed in aconitine-simulated ventricular fibrillation (P < 0.0001) and ouabain-simulated III° atrioventricular block (P < 0.0001). Shot-lasting electric shock, e.g., ≤ 40 s, decreased LPLs level, while long-lasting, e.g., 5 min, increased it (fold change = 2.27, P = 0.0008). LPLs accumulation was observed in long-lasting myocardial ischemia, e.g., 4 h (fold change = 1.20, P = 0.0012), when caspase3 activity was elevated (P = 0.0012), indicating increased cell death, but not coincided with higher frequent arrhythmias. In postmortem human ventricular myocardium, differences of LPLs level in left ventricular myocardium was not observed among coronary artery disease- and other heart diseases-caused sudden death and non-heart disease caused death. LPLs level manifested a remarkable increasing from postmortem 12 h on in rats, thus abolishing the potential for serving as biomarkers of sudden cardiac death. Token together, in this study we found that LPLs in ventricular myocardium were initially decreased by the onset of ischemia, LPLs accumulation do not confer arrhythmogenesis during acute myocardial ischemia. It is necessary to reassess the roles of LPLs in myocardial infarction.
Collapse
Affiliation(s)
- Jiawei Liu
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tingting Mai
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Han Ren
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yafei Chang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Chao Li
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Guoli Lv
- Guangzhou Forensic Science Institute, Guangzhou, 510030, China
- Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510050, China
| | - Da Zheng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xinbiao Liao
- Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510050, China
| | - Yangeng Yu
- Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510050, China
| | - Fu Zhang
- Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510050, China
| | - Shuquan Zhao
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaoshan Liu
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shuiping Liu
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hu Zhao
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Bin Luo
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Chao Liu
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510050, China.
- National Anti-Drug Laboratory Guangdong Regional Center, Guangzhou, 510230, China.
| | - Erwen Huang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
5
|
Barreto II, Gonçalves LR, Corrêa AF, Marin-Morales MA, Moraes KCM. Predictive toxicological effects of Artemisia absinthium essential oil on hepatic stellate cells. Toxicol In Vitro 2024; 95:105738. [PMID: 38000518 DOI: 10.1016/j.tiv.2023.105738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Medicinal plants are important worldwide, considering their properties for treating diseases; however, few studies have evaluated their toxicological potential. Among them, Artemisia absinthium is frequently used to treat liver diseases, because its essential oil has several popular therapeutic properties. Based on this information, in the present study, we investigated molecular connectors of physiological effects of the Artemisia absinthium essential oil on human hepatic stellate cell line, LX-2, to explore the potential toxicity of the plant on liver cells. LX-2 is a cellular model to investigate mechanisms of liver fibrosis; then, to analyze the essential oil effects LX-2 was cultured under different conditions, treated or not with the essential oil at 0.4 μg/μL for 24 h. Next, fluorescence microscopy analyses, gene expression measurements, and biochemical approaches revealed that the essential oil reduced pro-fibrogenic markers; however, disrupt lipid metabolism, and cause cellular stress, by the activation of cellular detoxification and pro-inflammatory processes. In conclusion, the hepatic stellate cells incubated with the essential oil present an antifibrotic potential, supporting its popular use; however, the combined results suggest that the essential oil of Artemisia absinthium should be used with caution.
Collapse
Affiliation(s)
- I I Barreto
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Programa de Pós-Graduação em Biotecnologia, Campus Araraquara, Instituto de Química, Araraquara, SP, Brazil; Laboratório de Sinalização Celular e Expressão Gênica, Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Rio Claro, SP, Brazil
| | - L R Gonçalves
- Laboratório de Mutagênese Ambiental, Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Rio Claro, SP, Brazil
| | - A F Corrêa
- Laboratório de Mutagênese Ambiental, Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Rio Claro, SP, Brazil
| | - M A Marin-Morales
- Laboratório de Mutagênese Ambiental, Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Rio Claro, SP, Brazil
| | - K C M Moraes
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Programa de Pós-Graduação em Biotecnologia, Campus Araraquara, Instituto de Química, Araraquara, SP, Brazil; Laboratório de Sinalização Celular e Expressão Gênica, Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Rio Claro, SP, Brazil.
| |
Collapse
|
6
|
Lin LC, Tu B, Song K, Liu ZY, Sun H, Zhou Y, Sha JM, Yang JJ, Zhang Y, Zhao JY, Tao H. Mitochondrial quality control in cardiac fibrosis: Epigenetic mechanisms and therapeutic strategies. Metabolism 2023:155626. [PMID: 37302693 DOI: 10.1016/j.metabol.2023.155626] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Cardiac fibrosis (CF) is considered an ultimate common pathway of a wide variety of heart diseases in response to diverse pathological and pathophysiological stimuli. Mitochondria are characterized as isolated organelles with a double-membrane structure, and they primarily contribute to and maintain highly dynamic energy and metabolic networks whose distribution and structure exert potent support for cellular properties and performance. Because the myocardium is a highly oxidative tissue with high energy demands to continuously pump blood, mitochondria are the most abundant organelles within mature cardiomyocytes, accounting for up to one-third of the total cell volume, and play an essential role in maintaining optimal performance of the heart. Mitochondrial quality control (MQC), including mitochondrial fusion, fission, mitophagy, mitochondrial biogenesis, and mitochondrial metabolism and biosynthesis, is crucial machinery that modulates cardiac cells and heart function by maintaining and regulating the morphological structure, function and lifespan of mitochondria. Certain investigations have focused on mitochondrial dynamics, including manipulating and maintaining the dynamic balance of energy demand and nutrient supply, and the resultant findings suggest that changes in mitochondrial morphology and function may contribute to bioenergetic adaptation during cardiac fibrosis and pathological remodeling. In this review, we discuss the function of epigenetic regulation and molecular mechanisms of MQC in the pathogenesis of CF and provide evidence for targeting MQC for CF. Finally, we discuss how these findings can be applied to improve the treatment and prevention of CF.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - He Sun
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Yang Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ji-Ming Sha
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Jing-Jing Yang
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
7
|
Jia W, Wu X, Shi L. Naturally forming benzoic acid orientates perilipin to facilitate glyceride-type polyunsaturated fatty acid degradation via fermentation behavior. J Dairy Sci 2023; 106:1650-1671. [PMID: 36710193 DOI: 10.3168/jds.2022-22381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/07/2022] [Indexed: 01/29/2023]
Abstract
Naturally forming benzoic acid in fermented dairy products accumulates in organisms and biomagnifies through collateral transport. The association between benzoic acid agglomeration and susceptible lipid nutrients remains obscure. Horizontal analysis of lipidomic alteration in response to benzoic acid was conducted and the spatially proteomic map was constructed using label-free quantitative proteomics. From synergistic integration of multi-omics in benzoic acid accumulated fermented goat milk model, the biological processes of significant proteins mostly focused on glyceride-type polyunsaturated fatty acids degradation (143.818 ± 0.51 mg/kg to 104.613 ± 0.29 mg/kg). As a physiological barrier shield, perilipin, which is coated on the surface of lipid droplets, protects triacylglycerols from cytosolic lipases, thus preventing triglyceride hydrolysis. The expression of perilipin decreased by 90% compared with the control group, leading to the decrease of triglycerides. Benzoic acid suppressed phosphatidylethanolamines and phosphatidylcholines synthesis by attenuating choline phosphotransferase and ethanolamine phosphotransferase. Less diglyceride generated by the dephosphorylation of phosphatidic acid entered choline phosphotransferase and ethanolamine phosphotransferase-mediated glycerophospholipid metabolisms. Fermentation of goat milk at a low temperature and less incubation time leads to the production of less benzoic acid and mitigation of lipid nutrient loss. The present study delineated the molecular landscape of fermented goat milk containing endogenous benzoic acid and further dissected the trajectory guiding lipid alteration to advance control of benzoic acid residue.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, 710021 China.
| | - Xixuan Wu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China
| | - Lin Shi
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021 China
| |
Collapse
|
8
|
Quiles JM, Gustafsson ÅB. The role of mitochondrial fission in cardiovascular health and disease. Nat Rev Cardiol 2022; 19:723-736. [PMID: 35523864 PMCID: PMC10584015 DOI: 10.1038/s41569-022-00703-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2022] [Indexed: 02/07/2023]
Abstract
Mitochondria are organelles involved in the regulation of various important cellular processes, ranging from ATP generation to immune activation. A healthy mitochondrial network is essential for cardiovascular function and adaptation to pathological stressors. Mitochondria undergo fission or fusion in response to various environmental cues, and these dynamic changes are vital for mitochondrial function and health. In particular, mitochondrial fission is closely coordinated with the cell cycle and is linked to changes in mitochondrial respiration and membrane permeability. Another key function of fission is the segregation of damaged mitochondrial components for degradation by mitochondrial autophagy (mitophagy). Mitochondrial fission is induced by the large GTPase dynamin-related protein 1 (DRP1) and is subject to sophisticated regulation. Activation requires various post-translational modifications of DRP1, actin polymerization and the involvement of other organelles such as the endoplasmic reticulum, Golgi apparatus and lysosomes. A decrease in mitochondrial fusion can also shift the balance towards mitochondrial fission. Although mitochondrial fission is necessary for cellular homeostasis, this process is often aberrantly activated in cardiovascular disease. Indeed, strong evidence exists that abnormal mitochondrial fission directly contributes to disease development. In this Review, we compare the physiological and pathophysiological roles of mitochondrial fission and discuss the therapeutic potential of preventing excessive mitochondrial fission in the heart and vasculature.
Collapse
Affiliation(s)
- Justin M Quiles
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Xie S, Zhang M, Shi W, Xing Y, Huang Y, Fang WX, Liu SQ, Chen MY, Zhang T, Chen S, Zeng X, Wang S, Deng W, Tang Q. Long-Term Activation of Glucagon-like peptide-1 receptor by Dulaglutide Prevents Diabetic Heart Failure and Metabolic Remodeling in Type 2 Diabetes. J Am Heart Assoc 2022; 11:e026728. [PMID: 36172969 DOI: 10.1161/jaha.122.026728] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Mechanistic insights of glucagon-like peptide-1 receptor agonists remain incompletely identified, despite the efficacy in heart failure observed in clinical trials. Here, we evaluated the effects of dulaglutide on heart complications and illuminated its underlying mechanism. Methods and Results We used mice with high-fat diet (HFD)/streptozotocin-induced type 2 diabetes to investigate the effects of dulaglutide upon diabetic cardiac dysfunction. After the onset of diabetes, control and diabetic mice were injected subcutaneously with either dulaglutide (type 2 diabetes-dulaglutide and control-dulaglutide groups) or vehicle (type 2 diabetes-vehicle and control-vehicle groups) for 8 weeks. Subsequently, heart characteristics, cardiometabolic profile and mitochondrial morphology and function were evaluated. Also, we analyzed the effects of dulaglutide on neonatal rat ventricular myocytes treated with high glucose plus palmitic acid. In addition, wild type and AMP-activated protein kinase α2 mutant mice were used to evaluate the underlying mechanism. In type 2 diabetes mouse model, dulaglutide ameliorated insulin resistance, improved glucose tolerance, reduced hyperlipidemia, and promoted fatty acid use in the myocardium. Dulaglutide treatment functionally attenuated cardiac remodeling and dysfunction and promoted metabolic reprogramming in diabetic mice. Furthermore, dulaglutide improved mitochondria fragmentation in myocytes, and simultaneously reinstated mitochondrial morphology and function in diabetic hearts. We also found that dulaglutide preserved AMP-activated protein kinase α2-dependent mitochondrial homeostasis, and the protective effects of dulaglutide on diabetic heart was almost abated by AMP-activated protein kinase α2 knockout. Conclusions Dulaglutide prevents diabetic heart failure and favorably affects myocardial metabolic remodeling by impeding mitochondria fragmentation, and we suggest a potential strategy to develop a long-term activation of glucagon-like peptide-1 receptor-based therapy to treat diabetes associated cardiovascular complications.
Collapse
Affiliation(s)
- Saiyang Xie
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Min Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Wenke Shi
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Yun Xing
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Yan Huang
- Department of Endocrinology Renmin Hospital of Wuhan University Wuhan P.R. China
| | - Wen-Xi Fang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Shi-Qiang Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Meng-Ya Chen
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Tong Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University Wuhan P.R. China
| | - Xiaofeng Zeng
- Cardiovascular Research Institute of Wuhan University Wuhan P.R. China
| | - Shasha Wang
- Cardiovascular Research Institute of Wuhan University Wuhan P.R. China
| | - Wei Deng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| | - Qizhu Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan P.R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan P.R. China
| |
Collapse
|
10
|
Zhou M, Li C, Han X, Yu B, Yan XZ, Zhang Y, Yang XJ. Lipidomic analysis reveals altered lipid profiles of gingival tissues with periodontitis. J Clin Periodontol 2022; 49:1192-1202. [PMID: 35924763 DOI: 10.1111/jcpe.13710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
Abstract
AIM The role of lipids in periodontitis has not been well studied. Thus, this study aimed to explore periodontitis-associated lipid profile changes and identify differentially expressed lipid metabolites in gingival tissues. MATERIALS AND METHODS Gingival tissues from 38 patients with periodontitis (periodontitis group) and 38 periodontally healthy individuals (control group) were collected. A UHPLC-QTOF-MS-based non-targeted metabolomics platform was used to identify and compare the lipid profiles of the two groups. The distribution and expression of related proteins were subsequently analyzed via immunohistochemistry to further validate the identified lipids. RESULTS Lipid profiles significantly differed between the two groups, and 20 differentially expressed lipid species were identified. Lysophosphatidylcholines (lysoPCs), diacylglycerols (DGs), and phosphatidylethanolamines (PEs) were significantly upregulated, while triacylglycerols (TGs) were downregulated in the periodontitis group. Moreover, the staining intensity of ABHD5/CGI-58, secretory phospholipase A2 (sPLA2), and sPLA2-IIA was significantly stronger in the gingival tissues of patients with periodontitis than in those of healthy controls. CONCLUSIONS LysoPCs, DGs, and PEs were significantly upregulated, whereas TGs were downregulated in gingival tissues of patients with periodontitis. Correspondingly, the immunohistochemical staining of ABHD5/CGI-58, sPLA2, and sPLA2-IIA in gingival tissues was consistent with the downstream production of lipid classes (lysoPCs, TGs, and DGs).
Collapse
Affiliation(s)
- Min Zhou
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Chen Li
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xue Han
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Bohan Yu
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiang-Zhen Yan
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yan Zhang
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiao-Juan Yang
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
11
|
Garvin AM, Hale TM. State of Change: Epigenetic and Mitochondrial Regulation of Cardiac Fibroblast Activation. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
12
|
Wichaiyo S, Saengklub N. Alterations of sodium-hydrogen exchanger 1 function in response to SGLT2 inhibitors: what is the evidence? Heart Fail Rev 2022; 27:1973-1990. [PMID: 35179683 DOI: 10.1007/s10741-022-10220-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
This review summarizes and describes the current evidence addressing how sodium-glucose cotransporter 2 (SGLT2) inhibitors alter the function of sodium-hydrogen exchanger 1 (NHE-1), in association with their protective effects against adverse cardiovascular events. In the heart, SGLT2 inhibitors modulate the function of NHE-1 (either by direct inhibition or indirect attenuation of protein expression), which promotes cardiac contraction and an enhanced energy supply, in association with improved mitochondrial function, reduced inflammation/oxidative/endoplasmic reticulum stress, and attenuated fibrosis and apoptotic/autophagic cell death. The vasodilating effect of SGLT2 inhibitors has also been proposed due to NHE-1 inhibition. Moreover, platelet-expressed NHE-1 might serve as a target for SGLT2 inhibitors, since these drugs and selective NHE-1 inhibitors produce comparable activity against adenosine diphosphate-stimulated platelet activation. Overall, it is promising that the modulation of the functions of NHE-1 on the heart, blood vessels, and platelets may act as a contributing pathway for the cardiovascular benefits of SGLT2 inhibitors in diabetes and heart failure.
Collapse
Affiliation(s)
- Surasak Wichaiyo
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Rajathevi, Bangkok, 10400, Thailand. .,Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Nakkawee Saengklub
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Cabrera JTO, Makino A. Efferocytosis of vascular cells in cardiovascular disease. Pharmacol Ther 2022; 229:107919. [PMID: 34171333 PMCID: PMC8695637 DOI: 10.1016/j.pharmthera.2021.107919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022]
Abstract
Cell death and the clearance of apoptotic cells are tightly regulated by various signaling molecules in order to maintain physiological tissue function and homeostasis. The phagocytic removal of apoptotic cells is known as the process of efferocytosis, and abnormal efferocytosis is linked to various health complications and diseases, such as cardiovascular disease, inflammatory diseases, and autoimmune diseases. During efferocytosis, phagocytic cells and/or apoptotic cells release signals, such as "find me" and "eat me" signals, to stimulate the phagocytic engulfment of apoptotic cells. Primary phagocytic cells are macrophages and dendritic cells; however, more recently, other neighboring cell types have also been shown to exhibit phagocytic character, including endothelial cells and fibroblasts, although they are comparatively slower in clearing dead cells. In this review, we focus on macrophage efferocytosis of vascular cells, such as endothelial cells, smooth muscle cells, fibroblasts, and pericytes, and its relation to the progression and development of cardiovascular disease. We also highlight the role of efferocytosis-related molecules and their contribution to the maintenance of vascular homeostasis.
Collapse
Affiliation(s)
- Jody Tori O Cabrera
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
14
|
Mitochondrial Metabolism in Carcinogenesis and Cancer Therapy. Cancers (Basel) 2021; 13:cancers13133311. [PMID: 34282749 PMCID: PMC8269082 DOI: 10.3390/cancers13133311] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Reprogramming metabolism is a hallmark of cancer. Warburg’s effect, defined as increased aerobic glycolysis at the expense of mitochondrial respiration in cancer cells, opened new avenues of research in the field of cancer. Later findings, however, have revealed that mitochondria remain functional and that they actively contribute to metabolic plasticity of cancer cells. Understanding the mechanisms by which mitochondrial metabolism controls tumor initiation and progression is necessary to better characterize the onset of carcinogenesis. These studies may ultimately lead to the design of novel anti-cancer strategies targeting mitochondrial functions. Abstract Carcinogenesis is a multi-step process that refers to transformation of a normal cell into a tumoral neoplastic cell. The mechanisms that promote tumor initiation, promotion and progression are varied, complex and remain to be understood. Studies have highlighted the involvement of oncogenic mutations, genomic instability and epigenetic alterations as well as metabolic reprogramming, in different processes of oncogenesis. However, the underlying mechanisms still have to be clarified. Mitochondria are central organelles at the crossroad of various energetic metabolisms. In addition to their pivotal roles in bioenergetic metabolism, they control redox homeostasis, biosynthesis of macromolecules and apoptotic signals, all of which are linked to carcinogenesis. In the present review, we discuss how mitochondria contribute to the initiation of carcinogenesis through gene mutations and production of oncometabolites, and how they promote tumor progression through the control of metabolic reprogramming and mitochondrial dynamics. Finally, we present mitochondrial metabolism as a promising target for the development of novel therapeutic strategies.
Collapse
|
15
|
Yang CM, Yang CC, Hsiao LD, Yu CY, Tseng HC, Hsu CK, Situmorang JH. Upregulation of COX-2 and PGE 2 Induced by TNF-α Mediated Through TNFR1/MitoROS/PKCα/P38 MAPK, JNK1/2/FoxO1 Cascade in Human Cardiac Fibroblasts. J Inflamm Res 2021; 14:2807-2824. [PMID: 34234507 PMCID: PMC8254141 DOI: 10.2147/jir.s313665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose Tumor necrosis factor-α (TNF-α) has been shown to exert as a pathogenic factor in cardiac fibrosis and heart failure which were associated with the up-regulation of cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) axis. However, whether TNF-α-induced COX-2/PGE2 upregulation mediated through ROS-dependent cascade remains elusive in human cardiac fibroblasts (HCFs). This study aims to address the underlying mechanisms of TNF-α-induced COX-2/PGE2 expression. Methods Here, we used TNF receptor neutralizing antibody (TNFR nAb), pharmacologic inhibitors, and siRNAs to dissect the involvement of signaling components examined by Western blot and ELISA in TNF-α-mediated responses in HCFs. MitoSOX Red was used to measure mitoROS generation. Isolation of subcellular fractions was performed to determine membrane translocation of PKCα. Promoter luciferase assay and chromatin immunoprecipitation (ChIP) assay were used to determine the role of transcription factor. Results We found that TNF-α time- and concentration-dependently upregulated COX-2 protein and mRNA expression as well as PGE2 synthesis which was attenuated by TNFR1 nAb, the inhibitor of mitochondrial ROS scavenger (MitoTEMPO), protein kinase C [(PKC)α, Gö6976], p38 MAPK [p38 inhibitor VIII, (p38i VIII)], JNK1/2 (SP600125), or forkhead box protein O1 [(FoxO1), AS1842856], and transfection with their respective siRNAs in HCFs. TNF-α-stimulated PKCα phosphorylation was inhibited by TNFR1 nAb, MitoTEMPO, or Gö6976. TNF-α stimulated phosphorylation of p38 MAPK and JNK1/2 was attenuated by TNFR1 nAb, MitoTEMPO, Gö6976, and their inhibitors p38i VIII and SP600125. Moreover, TNF-α-triggered FoxO1 phosphorylation was abolished by AS1842856, TNFR1 nAb, and its upstream inhibitors MitoTEMPO, Gö6976, p38i VIII, and SP600125. Phosphorylation of FoxO1 could enhance its interaction with the COX-2 promoter element revealed by ChIP assay, which was attenuated by AS1842856. Conclusion Our results suggested that TNF-α-induced COX-2/PGE2 upregulation is mediated through TNFR1-dependent MitoROS/PKCα/p38 MAPK and JNK1/2 cascade to activate FoxO1 binding with the COX-2 promoter in HCFs.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan.,Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, 40402, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung, 41354, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan, 33302, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Ying Yu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Hui-Ching Tseng
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chih-Kai Hsu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Jiro Hasegawa Situmorang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| |
Collapse
|
16
|
Zhao H, He Y. The Inhibitory Effect of Lysophosphatidylcholine on Proangiogenesis of Human CD34 + Cells Derived Endothelial Progenitor Cells. Front Mol Biosci 2021; 8:682367. [PMID: 34179086 PMCID: PMC8223510 DOI: 10.3389/fmolb.2021.682367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Increasing evidence reveals that lysophosphatidylcholine (LPC) is closely related to endothelial dysfunction. The present study aimed to investigate the mechanism of LPC in inhibiting the proangiogenesis and vascular inflammation of human endothelial progenitor cells (EPCs) derived from CD34+ cells. The early EPCs were derived from CD34+ hematopoietic stem cells whose purity was identified using flow cytometry analysis. The surface markers (CD34, KDR, CD31; VE-cadherin, vWF, eNOS) of EPCs were examined by flow cytometry analysis and immunofluorescence. RT-qPCR was used to detect the mRNA expression of inflammatory cytokines (CCL2, IL-8, CCL4) and genes associated with angiogenesis (VEGF, ANG-1, ANG-2) in early EPCs after treatment of LPC (10 μg/ml) or phosphatidylcholine (PC, 10 μg/ml, control). The angiogenesis of human umbilical vein endothelial cells (HUVECs) incubated with the supernatants of early EPCs was detected by a tube formation assay. The mRNA and protein levels of key factors on the PKC pathway (phosphorylated PKC, TGF-β1) were measured by RT-qPCR and western blot. The localization of PKC-β1 in EPCs was determined by immunofluorescence staining. We found that LPC suppressed the expression of CCL2, CCL4, ANG-1, ANG-2, promoted IL-8 expression and had no significant effects on VEGF expression in EPCs. EPCs promoted the angiogenesis of HUVECs, which was significantly inhibited by LPC treatment. Moreover, LPC was demonstrated to promote the activation of the PKC signaling pathway in EPCs. In conclusion, LPC inhibits proangiogenesis of human endothelial progenitor cells derived from CD34+ hematopoietic stem cells.
Collapse
Affiliation(s)
- Haijun Zhao
- Department of Pain, The First Hospital of Jilin University, Changchun, China
| | - Yanhui He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Iwata Y, Sakai N, Nakajima Y, Oshima M, Nakagawa-Yoneda S, Ogura H, Sato K, Minami T, Kitajima S, Toyama T, Yamamura Y, Miyagawa T, Hara A, Shimizu M, Furuichi K, Wada T. Anti-fibrotic potential of erythropoietin signaling on bone marrow derived fibrotic cell. BMC Nephrol 2021; 22:203. [PMID: 34059008 PMCID: PMC8167964 DOI: 10.1186/s12882-021-02411-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/13/2021] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The number of patients with end stage kidney disease (ESKD) are increasing world-side. While interstitial fibrosis (IF) is a common step for the progression to ESKD, therapeutic options for IF is still limited in clinical settings. We have reported that bone marrow-derived fibrotic cell, fibrocyte, is involved in the pathogenesis of kidney fibrosis. Also recent studies revealed that erythropoietin has protective effect on kidney diseases. However, it is unknown whether erythropoietin (EPO) inhibits fibrosis in progressive kidney injury. Therefore, we explored the impacts of EPO on kidney fibrosis with focusing on fibrocyte. METHOD Fibrocyte was differentiated from peripheral mononuclear cells of healthy donor. Fibrocyte was stimulated with transforming growth factor beta (TGF)-β with/without EPO treatment. Moreover, the therapeutic effect of EPO was evaluated in murine unilateral ureteral obstruction (UUO) model. RESULT TGF-β stimulation increased the expression of COL1 mRNA in fibrocyte. EPO signal reduced the expression of COL1 mRNA in dose dependent manner. EPO reduced mitochondrial oxidative stress and ameliorated mitochondrial membrane depolarization induced by TGF-β stimulation. Moreover, EPO reduced the mRNA expression of mitochondria related molecules, TRAF6, in fibrocyte. In addition, the count of CD45+/αSMA + double-positive fibrocyte was decreased in the EPO-administered UUO kidneys. CONCLUSION EPO signals function to prevent kidney fibrosis, particularly in fibrocyte. Regulating the renal accumulation of fibrocyte is a part of the anti-fibrotic functions of EPO.
Collapse
Affiliation(s)
- Yasunori Iwata
- Division of Infection Control, Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, 920-8641, Kanazawa , Japan. .,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan.
| | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Division of Blood Purification, Kanazawa University Hospital, Ishikawa, Kanazawa, Japan
| | - Yuki Nakajima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Megumi Oshima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | | | - Hisayuki Ogura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Koichi Sato
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Taichiro Minami
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Division of Blood Purification, Kanazawa University Hospital, Ishikawa, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Taro Miyagawa
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Kengo Furuichi
- Division of Nephrology, Kanazawa Medical University School of Medicine, Ishikawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
18
|
Onset of Senescence and Steatosis in Hepatocytes as a Consequence of a Shift in the Diacylglycerol/Ceramide Balance at the Plasma Membrane. Cells 2021; 10:cells10061278. [PMID: 34064003 PMCID: PMC8224046 DOI: 10.3390/cells10061278] [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: 03/30/2021] [Revised: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Ceramide and diacylglycerol (DAG) are bioactive lipids and mediate many cellular signaling pathways. Sphingomyelin synthase (SMS) is the single metabolic link between the two, while SMS2 is the only SMS form located at the plasma membrane. SMS2 functions were investigated in HepG2 cell lines stably expressing SMS2. SMS2 overexpression did not alter sphingomyelin (SM), phosphatidylcholine (PC), or ceramide levels. DAG content increased by approx. 40% and led to downregulation of DAG-dependent protein kinase C (PKC). SMS2 overexpression also induced senescence, characterized by positivity for β-galactosidase activity and heterochromatin foci. HepG2-SMS2 cells exhibited protruded mitochondria and suppressed mitochondrial respiration rates. ATP production and the abundance of Complex V were substantially lower in HepG2-SMS2 cells as compared to controls. SMS2 overexpression was associated with inflammasome activation based on increases in IL-1β and nlpr3 mRNA levels. HepG2-SMS2 cells exhibited lipid droplet accumulation, constitutive activation of AMPK based on elevated 172Thr phosphorylation, increased AMPK abundance, and insensitivity to insulin suppression of AMPK. Thus, our results show that SMS2 regulates DAG homeostasis and signaling in hepatocytes and also provide proof of principle for the concept that offset in bioactive lipids’ production at the plasma membrane can drive the senescence program in association with steatosis and, seemingly, by cell-autonomous mechanisms.
Collapse
|
19
|
Gao QY, Zhang HF, Tao J, Chen ZT, Liu CY, Liu WH, Wu MX, Yin WY, Gao GH, Xie Y, Yang Y, Liu PM, Wang JF, Chen YX. Mitochondrial Fission and Mitophagy Reciprocally Orchestrate Cardiac Fibroblasts Activation. Front Cell Dev Biol 2021; 8:629397. [PMID: 33585469 PMCID: PMC7874126 DOI: 10.3389/fcell.2020.629397] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022] Open
Abstract
Although mitochondrial fission has been reported to increase proliferative capacity and collagen production, it can also contribute to mitochondrial impairment, which is detrimental to cell survival. The aim of the present study was to investigate the role of mitochondrial fission in cardiac fibroblasts (CF) activation and explore the mechanisms involved in the maintenance of mitochondrial health under this condition. For this, changes in the levels of mitochondrial fission/fusion-related proteins were assessed in transforming growth factor beta 1 (TGF-β1)-activated CF, whereas the role of mitochondrial fission during this process was also elucidated, as were the underlying mechanisms. The interaction between mitochondrial fission and mitophagy, the main defense mechanism against mitochondrial impairment, was also explored. The results showed that the mitochondria in TGF-β1-treated CF were noticeably more fragmented than those of controls. The expression of several mitochondrial fission-related proteins was markedly upregulated, and the levels of fusion-related proteins were also altered, but to a lesser extent. Inhibiting mitochondrial fission resulted in a marked attenuation of TGF-β1-induced CF activation. The TGF-β1-induced increase in glycolysis was greatly suppressed in the presence of a mitochondrial inhibitor, whereas a glycolysis-specific antagonist exerted little additional antifibrotic effects. TGF-β1 treatment increased cellular levels of reactive oxygen species (ROS) and triggered mitophagy, but this effect was reversed following the application of ROS scavengers. For the signals mediating mitophagy, the expression of Pink1, but not Bnip3l/Nix or Fundc1, exhibited the most significant changes, which could be counteracted by treatment with a mitochondrial fission inhibitor. Pink1 knockdown suppressed CF activation and mitochondrial fission, which was accompanied by increased CF apoptosis. In conclusion, mitochondrial fission resulted in increased glycolysis and played a crucial role in CF activation. Moreover, mitochondrial fission promoted reactive oxygen species (ROS) production, leading to mitophagy and the consequent degradation of the impaired mitochondria, thus promoting CF survival and maintaining their activation.
Collapse
Affiliation(s)
- Qing-Yuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Chi-Yu Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Wen-Hao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Wen-Yao Yin
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Guang-Hao Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Yong Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Ying Yang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Pin-Ming Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Yang-Xin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| |
Collapse
|
20
|
Song D, Xu C, Holck AL, Liu R. Acrylamide inhibits autophagy, induces apoptosis and alters cellular metabolic profiles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111543. [PMID: 33396091 DOI: 10.1016/j.ecoenv.2020.111543] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 06/12/2023]
Abstract
Acrylamide (ACR) is generated during thermal processing of carbohydrate-rich foods at high temperature and can directly enter the body through ingestion, inhalation and skin contact. The toxicity of ACR has been widely studied. The main results of these studies show that exposure to ACR can cause neurotoxicity in both animals and humans, and show reproductive toxicity and carcinogenicity in rodent animal models. However, the mechanism of toxicity of ACR has not been studied by metabolomics approaches, and the effect of ACR on autophagy remains unknown. Here, U2OS cell were treated with ACR 6 and 24 h and collected for further study. We have demonstrated that ACR inhibited autophagic flux, and increased ROS content. Accumulation of ROS resulted in increase of apoptosis rates and secretion of inflammatory factors. In addition, significant differences in metabolic profiles were observed between ACR treated and control cells according to multiple analysis models. A total of 73 key differential metabolites were identified. They were involved in multiple metabolic pathways. Among them, exposure to ACR caused glycolysis/gluconeogenesis attenuation by decreasing levels of glycolytic intermediates, reduced the rate of the TCA cycle, while elevating levels of several amino acid metabolites and lipid metabolites. In summary, our study provides useful evidence of cytotoxicity caused by ACR via metabolomics and multiple bioanalytic methods.
Collapse
Affiliation(s)
- Dan Song
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China; College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Chao Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Askild L Holck
- Norwegian Institute of Food, Fisheries and Aquaculture Research (NOFIMA), P.O. Box 210, N-1431 Aas, Norway
| | - Rong Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China; National center for international research on animal gut nutrition, Nanjing, China; Jiangsu collaborative innovation center of meat production and processing, Nanjing, China.
| |
Collapse
|
21
|
Narzt MS, Pils V, Kremslehner C, Nagelreiter IM, Schosserer M, Bessonova E, Bayer A, Reifschneider R, Terlecki-Zaniewicz L, Waidhofer-Söllner P, Mildner M, Tschachler E, Cavinato M, Wedel S, Jansen-Dürr P, Nanic L, Rubelj I, El-Ghalbzouri A, Zoratto S, Marchetti-Deschmann M, Grillari J, Gruber F, Lämmermann I. Epilipidomics of Senescent Dermal Fibroblasts Identify Lysophosphatidylcholines as Pleiotropic Senescence-Associated Secretory Phenotype (SASP) Factors. J Invest Dermatol 2020; 141:993-1006.e15. [PMID: 33333126 DOI: 10.1016/j.jid.2020.11.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
During aging, skin accumulates senescent cells. The transient presence of senescent cells, followed by their clearance by the immune system, is important in tissue repair and homeostasis. The persistence of senescent cells that evade clearance contributes to the age-related deterioration of the skin. The senescence-associated secretory phenotype of these cells contains immunomodulatory molecules that facilitate clearance but also promote chronic damage. Here, we investigated the epilipidome-the oxidative modifications of phospholipids-of senescent dermal fibroblasts, because these molecules are among the bioactive lipids that were recently identified as senescence-associated secretory phenotype factors. Using replicative- and stress- induced senescence protocols, we identified lysophosphatidylcholines as universally elevated in senescent fibroblasts, whereas other oxidized lipids displayed a pattern that was characteristic for the used senescence protocol. When we tested the lysophosphatidylcholines for senescence-associated secretory phenotype activity, we found that they elicit chemokine release in nonsenescent fibroblasts but also interfere with toll-like receptor 2 and 6/CD36 signaling and phagocytic capacity in macrophages. Using matrix-assisted laser desorption/ionization Fourier transform ion cyclotron resonance mass spectrometry imaging, we localized two lysophosphatidylcholine species in aged skin. This suggests that lysophospholipids may facilitate immune evasion and low-grade chronic inflammation in skin aging.
Collapse
Affiliation(s)
- Marie-Sophie Narzt
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria
| | - Vera Pils
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Christopher Kremslehner
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria
| | - Ionela-Mariana Nagelreiter
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, Vienna, Austria
| | - Markus Schosserer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Emilia Bessonova
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Alina Bayer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Raffaela Reifschneider
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Petra Waidhofer-Söllner
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Cavinato
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Sophia Wedel
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Lucia Nanic
- Ruder Boskovic Institute, Division of Molecular Biology, Laboratory for Molecular and Cellular Biology, Zagreb, Croatia
| | - Ivica Rubelj
- Ruder Boskovic Institute, Division of Molecular Biology, Laboratory for Molecular and Cellular Biology, Zagreb, Croatia
| | | | - Samuele Zoratto
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Martina Marchetti-Deschmann
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Florian Gruber
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria.
| | - Ingo Lämmermann
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| |
Collapse
|
22
|
Baghaki S, Yalcin CE, Baghaki HS, Aydin SY, Daghan B, Yavuz E. COX2 inhibition in the treatment of COVID-19: Review of literature to propose repositioning of celecoxib for randomized controlled studies. Int J Infect Dis 2020; 101:29-32. [PMID: 33007455 PMCID: PMC7525269 DOI: 10.1016/j.ijid.2020.09.1466] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Coronavirus-triggered pulmonary and systemic disease, i.e. systemic inflammatory response to virally triggered lung injury, named COVID-19, and ongoing discussions on refining immunomodulation in COVID-19 without COX2 inhibition prompted us to search the related literature to show a potential target (COX2) and a weapon (celecoxib). The concept of selectively targeting COX2 and closely related cascades might be worth trying in the treatment of COVID-19 given the substantial amount of data showing that COX2, p38 MAPK, IL-1b, IL-6 and TGF-β play pivotal roles in coronavirus-related cell death, cytokine storm and pulmonary interstitial fibrosis. Considering the lack of definitive treatment and importance of immunomodulation in COVID-19, COX2 inhibition might be a valuable adjunct to still-evolving treatment strategies. Celecoxib has properties that should be evaluated in randomized controlled studies and is also available for off-label use.
Collapse
Affiliation(s)
- Semih Baghaki
- Istanbul University Cerrahpasa - Cerrahpasa Medical Faculty, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey.
| | - Can Ege Yalcin
- Istanbul University Cerrahpasa - Cerrahpasa Medical Faculty, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Hayriye Sema Baghaki
- Bakirkoy Sadi Konuk Research and Training Hospital, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Servet Yekta Aydin
- Istanbul University Cerrahpasa - Cerrahpasa Medical Faculty, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Basak Daghan
- Istanbul University Cerrahpasa - Cerrahpasa Medical Faculty, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| | - Ersin Yavuz
- Istanbul University Cerrahpasa - Cerrahpasa Medical Faculty, Department of Plastic, Reconstructive and Aesthetic Surgery, Istanbul, Turkey
| |
Collapse
|
23
|
Zanotti I, Marando S, Remaggi G, Bergonzi C, Bernini F, Bettini R, Elviri L. The adaptation of lipid profile of human fibroblasts to alginate 2D films and 3D printed scaffolds. Biochim Biophys Acta Gen Subj 2020; 1865:129734. [PMID: 32956750 DOI: 10.1016/j.bbagen.2020.129734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND The investigation of the interactions between cells and active materials is pivotal in the emerging 3D printing-biomaterial application fields. Here, lipidomics has been used to explore the early impact of alginate (ALG) hydrogel architecture (2D films or 3D printed scaffolds) and the type of gelling agent (CaCl2 or FeCl3) on the lipid profile of human fibroblasts. METHODS 2D and 3D ALG scaffolds were prepared and characterized in terms of water content, swelling, mechanical resistance and morphology before human fibroblast seeding (8 days). Using a liquid chromatography-triple quadrupole-tandem mass spectrometry approach, selected ceramides (CER), lysophosphatidylcholines (LPC), lysophosphatidic acids (LPA) and free fatty acids (FFA) were analyzed. RESULTS The results showed a clear alteration in the CER expression profile depending of both the geometry and the gelling agent used to prepare the hydrogels. As for LPCs, the main parameter affecting their distribution is the scaffold architecture with a significant decrease in the relative expression levels of the species with higher chain length (C20 to C22) for 3D scaffolds compared to 2D films. In the case of FFAs and LPAs only slight differences were observed as a function of scaffold geometry or gelling agent. CONCLUSIONS Variations in the cell membrane lipid profile were observed for 3D cell cultures compared to 2D and these data are consistent with activation processes occurring through the mutual interactions between fibroblasts and ALG support. These unknown physiologically relevant changes add insights into the discussion about the relationship between biomaterial and the variations of cell biological functions.
Collapse
Affiliation(s)
- Ilaria Zanotti
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Silvia Marando
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Giulia Remaggi
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Carlo Bergonzi
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Franco Bernini
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Ruggero Bettini
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Lisa Elviri
- Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy.
| |
Collapse
|
24
|
Huang JP, Cheng ML, Wang CH, Huang SS, Hsieh PS, Chang CC, Kuo CY, Chen KH, Hung LM. Therapeutic potential of cPLA2 inhibitor to counteract dilated-cardiomyopathy in cholesterol-treated H9C2 cardiomyocyte and MUNO rat. Pharmacol Res 2020; 160:105201. [PMID: 32942017 DOI: 10.1016/j.phrs.2020.105201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE The pathogenesis of cardiomyopathy in metabolically unhealthy obesity (MUO) has been well studied. However, the pathogenesis of cardiomyopathy typically associated with high cholesterol levels in metabolically unhealthy nonobesity (MUNO) remains unclear. We investigated whether cholesterol-generated LysoPCs contribute to cardiomyopathy and the role of cytosolic phospholipase A2 (cPLA2) inhibitor in cholesterol-induced MUNO. EXPERIMENTAL APPROACH Cholesterol diet was performed in Sprague-Dawley rats that were fed either regular chow (C), or high cholesterol chow (HC), or HC diet with 10 % fructose in drinking water (HCF) for 12 weeks. LysoPCs levels were subsequently measured in rats and in MUNO human patients. The effects of cholesterol-mediated LysoPCs on cardiac injury, and the action of cPLA2 inhibitor, AACOCF3, were further assessed in H9C2 cardiomyocytes. KEY RESULTS HC and HCF rats fed cholesterol diets demonstrated a MUNO-phenotype and cholesterol-induced dilated cardiomyopathy (DCM). Upregulated levels of LysoPCs were found in rat myocardium and the plasma in MUNO human patients. Further testing in H9C2 cardiomyocytes revealed that cholesterol-induced atrophy and death of cardiomyocytes was due to mitochondrial dysfunction and conditions favoring DCM (i.e. reduced mRNA expression of ANF, BNP, DSP, and atrogin-1), and that AACOCF3 counteracted the cholesterol-induced DCM phenotype. CONCLUSION AND IMPLICATIONS Cholesterol-induced MUNO-DCM phenotype was counteracted by cPLA2 inhibitor, which is potentially useful for the treatment of LysoPCs-associated DCM in MUNO.
Collapse
Affiliation(s)
- Jiung-Pang Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Mei-Ling Cheng
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Chao-Hung Wang
- Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.
| | - Shiang-Suo Huang
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan.
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| | - Chih-Chun Chang
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei, Taiwan.
| | - Chao-Yu Kuo
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Kuan-Hsing Chen
- Kidney Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Li-Man Hung
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Kidney Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
25
|
Bahia PK, Hadley SH, Barannikov I, Sowells I, Kim SH, Taylor-Clark TE. Antimycin A increases bronchopulmonary C-fiber excitability via protein kinase C alpha. Respir Physiol Neurobiol 2020; 278:103446. [PMID: 32360368 DOI: 10.1016/j.resp.2020.103446] [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: 03/11/2020] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/17/2022]
Abstract
Inflammation can increase the excitability of bronchopulmonary C-fibers leading to excessive sensations and reflexes (e.g. wheeze and cough). We have previously shown modulation of peripheral nerve terminal mitochondria by antimycin A causes hyperexcitability in TRPV1-expressing bronchopulmonary C-fibers through the activation of protein kinase C (PKC). Here, we have investigated the PKC isoform responsible for this signaling. We found PKCβ1, PKCδ and PKCε were expressed by many vagal neurons, with PKCα and PKCβ2 expressed by subsets of vagal neurons. In dissociated vagal neurons, antimycin A caused translocation of PKCα but not the other isoforms, and only in TRPV1-lineage neurons. In bronchopulmonary C-fiber recordings, antimycin A increased the number of action potentials evoked by α,β-methylene ATP. Selective inhibition of PKCα, PKCβ1 and PKCβ2 with 50 nM bisindolylmaleimide I prevented the antimycin-induced bronchopulmonary C-fiber hyperexcitability, whereas selective inhibition of only PKCβ1 and PKCβ2 with 50 nM LY333531 had no effect. We therefore conclude that PKCα is required for antimycin-induced increases in bronchopulmonary C-fiber excitability.
Collapse
Affiliation(s)
- Parmvir K Bahia
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Stephen H Hadley
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ivan Barannikov
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Isobel Sowells
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Seol-Hee Kim
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|