1
|
Lu R, Cai H, Liu Y, Ma G, Wang J, Yan M, Zhang Z, Yu B, Li Z, Fang S. Long non-coding RNA AK023617 orchestrates atherosclerosis by regulating the circadian rhythm of immunity-related GTPase family M protein in macrophages. Noncoding RNA Res 2025; 11:262-272. [PMID: 39902258 PMCID: PMC11788686 DOI: 10.1016/j.ncrna.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/08/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Acute coronary events show a diurnal rhythm, and atherosclerotic plaque vulnerability, as a histomorphological characteristic of major adverse cardiovascular events, is a key target for intervention. Although oscillating microRNAs reduce plaque stability by facilitating macrophage apoptosis in lesions, whether rhythmic long non-coding RNA (lncRNA) can regulate diurnal oscillations in plaque stability and the potential underlying mechanism remain unclear. In this study, we examined whether rhythmic lncRNAs are involved in the pathogenesis and progression of atherosclerosis and detected a novel circadian lncRNA-AK023617, which is positively correlated with the peak occurrence of major adverse cardiovascular events. Transfection of short interfering RNA specific to lnc-AK023617 into THP-1 cells dampened the oscillation of immunity-related GTPase family M protein 1 (Irgm1), which is negatively related to plaque stability. In ApoE-/- mice fed a high-fat diet for 12 weeks, diurnal variations in lncAK023617 were consistent with the proportions of necroptotic cells in atherosclerotic plaques. In addition, reduced expression of lncAK023617 inhibited P-RIP3 and P-MLKL in THP-1 cells. Mechanistically, lncAK023617 interacted with the core molecular clock Bmal1 and promoted nuclear translocation of Bmal1, which could directly bind to the E-BOX elements in the Irgm1 promoter. Thus, oscillating lncAK023617 in macrophages can affect plaque stability by regulating necroptosis, which regulates circadian expression of the target gene Irgm1 by increasing the transcriptional activity of Bmal1, ultimately determining the diurnal oscillations in plaque stability. Therefore, lncAK023617 may serve as a specific target to ameliorate atherosclerotic plaque vulnerability.
Collapse
Affiliation(s)
- Rongzhe Lu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hengxuan Cai
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yige Liu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guanpeng Ma
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jiaxin Wang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Miao Yan
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhenming Zhang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Bo Yu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhaoying Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
2
|
Wu H, Gao W, Ma Y, Zhong X, Qian J, Huang D, Ge J. TRIM25-mediated XRCC1 ubiquitination accelerates atherosclerosis by inducing macrophage M1 polarization and programmed death. Inflamm Res 2024; 73:1445-1458. [PMID: 38896288 DOI: 10.1007/s00011-024-01906-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Macrophage-mediated cleaning up of dead cells is a crucial determinant in reducing coronary artery inflammation and maintaining vascular homeostasis. However, this process also leads to programmed death of macrophages. So far, the role of macrophage death in the progression of atherosclerosis remains controversial. Also, the underlying mechanism by which transcriptional regulation and reprogramming triggered by macrophage death pathways lead to changes in vascular inflammation and remodeling are still largely unknown. TRIM25-mediated RIG-I signaling plays a key role in regulation of macrophages fate, however the role of TRIM25 in macrophage death-mediated atherosclerotic progression remains unclear. This study aims to investigate the relationship between TRIM25 and macrophage death in atherosclerosis. METHODS A total of 34 blood samples of patients with coronary stent implantation, including chronic total occlusion (CTO) leisions (n = 14) or with more than 50% stenosis of a coronary artery but without CTO leisions (n = 20), were collected, and the serum level of TRIM25 was detected by ELISA. Apoe-/- mice with or without TRIM25 gene deletion were fed with the high-fat diet (HFD) for 12 weeks and the plaque areas, necrotic core size, aortic fibrosis and inflammation were investigated. TRIM25 wild-type and deficient macrophages were isolated, cultured and stimulated with ox-LDL, RNA-seq, real-time PCR, western blot and FACS experiments were used to screen and validate signaling pathways caused by TRIM25 deletion. RESULTS Downregulation of TRIM25 was observed in circulating blood of CTO patients and also in HFD-induced mouse aortas. After HFD for 12 weeks, TRIM25-/-ApoeE-/- mice developed smaller atherosclerotic plaques, less inflammation, lower collagen content and aortic fibrosis compared with TRIM25+/+ApoeE-/- mice. By RNA-seq and KEGG enrichment analysis, we revealed that deletion of TRIM25 mainly affected pyroptosis and necroptosis pathways in ox-LDL-induced macrophages, and the expressions of PARP1 and RIPK3, were significantly decreased in TRIM25 deficient macrophages. Overexpression of TRIM25 promoted M1 polarization and necroptosis of macrophages, while inhibition of PARP1 reversed this process. Further, we observed that XRCC1, a repairer of DNA damage, was significantly upregulated in TRIM25 deficient macrophages, inhibiting PARP1 activity and PARP1-mediated pro-inflammatory change, M1 polarization and necroptosis of macrophages. By contrast, TRIM25 overexpression mediated ubiquitination of XRCC1, and the inhibition of XRCC1 released PARP1, and activated macrophage M1 polarization and necroptosis, which accelerated aortic inflammation and atherosclerotic plaque progression. CONCLUSIONS Our study has uncovered a crucial role of the TRIM25-XRCC1Ub-PARP1-RIPK3 axis in regulating macrophage death during atherosclerosis, and we highlight the potential therapeutic significance of macrophage reprogramming regulation in preventing the development of atherosclerosis.
Collapse
Affiliation(s)
- Hongxian Wu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Wei Gao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Yuanji Ma
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Dong Huang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
4
|
Liang JY, Gao S, Jiang JM, Zhang P, Zou W, Tang XQ, Tang YY. Itaconate inhibits corticosterone-induced necroptosis and neuroinflammation via up-regulating menin in HT22 cells. J Physiol Biochem 2024; 80:393-405. [PMID: 38427168 DOI: 10.1007/s13105-024-01012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Corticosterone (CORT) damages hippocampal neurons as well as induces neuroinflammation. The tricarboxylic acid cycle metabolite itaconate has an anti-inflammatory role. Necroptosis is a form of programmed cell death, also known as inflammatory cell death. Menin is a multifunctional scaffold protein, which deficiency aggravates neuroinflammation. In this study, we explored whether itaconate inhibits CORT-induced neuroinflammation as well as necroptosis and further investigated the mediatory role of Menin in this protective effect of itaconate by using an exposure of CORT to HT22 cells (a hippocampal neuronal cell line). The viability of HT22 cells was examined by the cell counting kit 8 (CCK-8). The morphology of HT22 cells was observed by transmission electron microscope (TEM). The expressions of necroptosis-related proteins (p-RIP1/RIP1, p-RIP3/RIP3, and p-MLKL/MLKL) were evaluated by western blotting. The contents of inflammatory factors were detected by an enzyme-linked immunosorbent assay (ELISA) kit. Our results showed that CORT increases the contents of pro-inflammatory factors (IL-1β, TNF-α) as well as decreases the contents of anti-inflammatory factors (IL-4, IL-10) in HT22 cells. We also found that CORT increases the expressions of necroptosis-related proteins (p-RIP1/RIP1, p-RIP3/RIP3, and p-MLKL/MLKL) and decreases the cell viability in HT22 cells, indicating that CORT induces necroptosis in HT22 cells. Itaconate improves CORT-induced neuroinflammation and necroptosis. Furthermore, itaconate upregulates the expression of Menin in CORT-exposed HT22 cells. Importantly, silencing Menin abolishes the antagonistic effect of itaconate on CORT-induced necroptosis and neuroinflammation. In brief, these results indicated that itaconate protects HT22 cells against CORT-induced neuroinflammation and necroptosis via upregulating Menin.
Collapse
Affiliation(s)
- Jin-Yu Liang
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Shan Gao
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Jia-Mei Jiang
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Pin Zhang
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, No. 336 S Dongfeng Road, Hengyang, 421002, Hunan Province, People's Republic of China
| | - Wei Zou
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, No. 336 S Dongfeng Road, Hengyang, 421002, Hunan Province, People's Republic of China
| | - Xiao-Qing Tang
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China.
- Department of Neurology, The Second Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Yi-Yun Tang
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
5
|
Makuch M, Stepanechko M, Bzowska M. The dance of macrophage death: the interplay between the inevitable and the microenvironment. Front Immunol 2024; 15:1330461. [PMID: 38576612 PMCID: PMC10993711 DOI: 10.3389/fimmu.2024.1330461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024] Open
Abstract
Macrophages are highly plastic cells ubiquitous in various tissues, where they perform diverse functions. They participate in the response to pathogen invasion and inflammation resolution following the immune response, as well as the maintenance of homeostasis and proper tissue functions. Macrophages are generally considered long-lived cells with relatively strong resistance to numerous cytotoxic factors. On the other hand, their death seems to be one of the principal mechanisms by which macrophages perform their physiological functions or can contribute to the development of certain diseases. In this review, we scrutinize three distinct pro-inflammatory programmed cell death pathways - pyroptosis, necroptosis, and ferroptosis - occurring in macrophages under specific circumstances, and explain how these cells appear to undergo dynamic yet not always final changes before ultimately dying. We achieve that by examining the interconnectivity of these cell death types, which in macrophages seem to create a coordinated and flexible system responding to the microenvironment. Finally, we discuss the complexity and consequences of pyroptotic, necroptotic, and ferroptotic pathway induction in macrophages under two pathological conditions - atherosclerosis and cancer. We summarize damage-associated molecular patterns (DAMPs) along with other microenvironmental factors, macrophage polarization states, associated mechanisms as well as general outcomes, as such a comprehensive look at these correlations may point out the proper methodologies and potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Małgorzata Bzowska
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
6
|
Zhang J, Qian J, Zhang W, Chen X. The pathophysiological role of receptor-interacting protein kinase 3 in cardiovascular disease. Biomed Pharmacother 2023; 165:114696. [PMID: 37329707 DOI: 10.1016/j.biopha.2023.114696] [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: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 06/19/2023] Open
Abstract
Recent studies have found that receptor interacting protein kinase 3 (RIPK3) can mediate CaMK Ⅱ phosphorylation and oxidation, open mitochondrial permeability transition pore (mPTP), and induce myocardial necroptosis. The increased expression or phosphorylation of RIPK3 is one of the important markers of necroptosis; Inhibition of CaMK Ⅱ phosphorylation or oxidation significantly reduces RIPK3 mediated myocardial necroptosis; Studies have shown that necroptosis plays an important role in the occurrence and development of cardiovascular diseases; Using the selective inhibitor GSK '872 of RIPK3 can effectively inhibit the occurrence and development of cardiovascular diseases, and can reverse cardiovascular and cardiac dysfunction caused by overexpression of RIPK3. In this review, we provide a brief overview of the current knowledge on RIPK3 in mediating necroptosis, inflammatory response, and oxidative stress, and discussed the role of RIPK3 in cardiovascular diseases such as atherosclerosis, myocardial ischaemia, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, China
| | - Jianan Qian
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China
| | - Wei Zhang
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, China; School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China.
| | - Xianfen Chen
- Department of Pharmacy, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
7
|
Wu G, Yu G, Zheng M, Peng W, Li L. Recent Advances for Dynamic-Based Therapy of Atherosclerosis. Int J Nanomedicine 2023; 18:3851-3878. [PMID: 37469455 PMCID: PMC10352141 DOI: 10.2147/ijn.s402678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 05/06/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease, which may lead to high morbidity and mortality. Currently, the clinical treatment strategy for AS is administering drugs and performing surgery. However, advanced therapy strategies are urgently required because of the deficient therapeutic effects of current managements. Increased number of energy conversion-based organic or inorganic materials has been used in cancer and other major disease treatments, bringing hope to patients with the development of nanomedicine and materials. These treatment strategies employ specific nanomaterials with specific own physiochemical properties (external stimuli: light or ultrasound) to promote foam cell apoptosis and cholesterol efflux. Based on the pathological characteristics of vulnerable plaques, energy conversion-based nano-therapy has attracted increasing attention in the field of anti-atherosclerosis. Therefore, this review focuses on recent advances in energy conversion-based treatments. In addition to summarizing the therapeutic effects of various techniques, the regulated pathological processes are highlighted. Finally, the challenges and prospects for further development of dynamic treatment for AS are discussed.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| | - Guanye Yu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Meiling Zheng
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 101121, People’s Republic of China
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Lei Li
- National Clinical Research Center for Obstetric & Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People’s Republic of China
| |
Collapse
|
8
|
Yang F, Xu M, Chen X, Luo Y. Spotlight on porphyrins: Classifications, mechanisms and medical applications. Biomed Pharmacother 2023; 164:114933. [PMID: 37236030 DOI: 10.1016/j.biopha.2023.114933] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Photodynamic therapy (PDT) and sonodynamic therapy (SDT) are non-invasive treatment methods with obvious inhibitory effect on tumors and have few side effects, which have been widely concerned and explored by researchers. Sensitizer is the main factor in determining the therapeutic effect of PDT and SDT. Porphyrins, a group of organic compounds widespread in nature, can be activated by light or ultrasound and produce reactive oxygen species. Therefore, porphyrins as sensitizers in PDT have been widely explored and investigated for many years. Herein, we summarize the classical porphyrin compounds and their applications and mechanisms in PDT and SDT. The application of porphyrin in clinical diagnosis and imaging is also discussed. In conclusion, porphyrins have good application prospects in disease treatment as an important part of PDT or SDT, and in clinical diagnosis and imaging.
Collapse
Affiliation(s)
- Fuyu Yang
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China
| | - Meiqi Xu
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China
| | - Xiaoyu Chen
- Department of Neonatal, The Fourth Hospital of Harbin Medical University, Harbin
| | - Yakun Luo
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
9
|
Okamura K, Inoue H, Tanaka K, Ikematsu Y, Furukawa R, Ota K, Yoneshima Y, Iwama E, Okamoto I. Immunostimulatory oncolytic activity of coxsackievirus A11 in human malignant pleural mesothelioma. Cancer Sci 2023; 114:1095-1107. [PMID: 36369966 PMCID: PMC9986072 DOI: 10.1111/cas.15645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/14/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive solid cancer with a poor prognosis, whereas coxsackievirus A11 (CVA11) is a potential oncolytic virus for cancer treatment. We here investigated the oncolytic activity of CVA11 with human MPM cell lines. CVA11 infection was cytotoxic in all six MPM cell lines examined and showed no or minimal cytotoxicity toward normal human normal cell lines. MPM cells with a higher surface level of intercellular adhesion molecule-1 (ICAM-1) expression tended to be more susceptible to CVA11-induced cytotoxicity, and a neutralizing antibody to ICAM-1 attenuated such cytotoxicity. CVA11 infection activated signaling by Akt and extracellular signal-regulated kinase (ERK) pathways, and inhibitors of such signaling also abrogated CVA11-mediated cytotoxicity. Furthermore, CVA11 infection-triggered multiple modes of tumor cell death including apoptosis, pyroptosis, and necroptosis, and such death was accompanied by the release or exposure of the proinflammatory cytokine interleukin-1β and damage-associated molecular patterns such as calreticulin, high-mobility group box-1, annexin A1, and heat shock protein 70, which are hallmarks of immunogenic cell death. Notably, in vivo treatment of human MPM xenografts with intratumoral CVA11 injection resulted in significant suppression of tumor growth in SCID mice, and all mice infected with CVA11 showed no significant change in body weight. Our findings collectively suggest that the oncolytic activity of CVA11 for MPM is dependent on ICAM-1 as a virus receptor, as well as on Akt and ERK signaling, and that oncolytic virotherapy with CVA11 is a promising treatment modality with immunostimulatory activity for human MPM.
Collapse
Affiliation(s)
- Koji Okamura
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Inoue
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Ikematsu
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, National Hospital Organization Omuta Hospital, Fukuoka, Japan
| | - Rie Furukawa
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Respiratory Medicine, Hamanomachi Hospital, Fukuoka, Japan
| | - Keiichi Ota
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Puylaert P, Zurek M, Rayner KJ, De Meyer GRY, Martinet W. Regulated Necrosis in Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:1283-1306. [PMID: 36134566 DOI: 10.1161/atvbaha.122.318177] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During atherosclerosis, lipid-rich plaques are formed in large- and medium-sized arteries, which can reduce blood flow to tissues. This situation becomes particularly precarious when a plaque develops an unstable phenotype and becomes prone to rupture. Despite advances in identifying and treating vulnerable plaques, the mortality rate and disability caused by such lesions remains the number one health threat in developed countries. Vulnerable, unstable plaques are characterized by a large necrotic core, implying a prominent role for necrotic cell death in atherosclerosis and plaque destabilization. Necrosis can occur accidentally or can be induced by tightly regulated pathways. Over the past decades, different forms of regulated necrosis, including necroptosis, ferroptosis, pyroptosis, and secondary necrosis, have been identified, and these may play an important role during atherogenesis. In this review, we describe several forms of necrosis that may occur in atherosclerosis and how pharmacological modulation of these pathways can stabilize vulnerable plaques. Moreover, some challenges of targeting necrosis in atherosclerosis such as the presence of multiple death-inducing stimuli in plaques and extensive cross-talk between necrosis pathways are discussed. A better understanding of the role of (regulated) necrosis in atherosclerosis and the mechanisms contributing to plaque destabilization may open doors to novel pharmacological strategies and will enable clinicians to tackle the residual cardiovascular risk that remains in many atherosclerosis patients.
Collapse
Affiliation(s)
- Pauline Puylaert
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Michelle Zurek
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology and Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, ON, Canada (K.J.R.).,University of Ottawa Heart Institute, ON, Canada (K.J.R.)
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Wim Martinet
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| |
Collapse
|
11
|
Jahanbani F, Maynard RD, Sing JC, Jahanbani S, Perrino JJ, Spacek DV, Davis RW, Snyder MP. Phenotypic characteristics of peripheral immune cells of Myalgic encephalomyelitis/chronic fatigue syndrome via transmission electron microscopy: A pilot study. PLoS One 2022; 17:e0272703. [PMID: 35943990 PMCID: PMC9362953 DOI: 10.1371/journal.pone.0272703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/25/2022] [Indexed: 01/06/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex chronic multi-systemic disease characterized by extreme fatigue that is not improved by rest, and worsens after exertion, whether physical or mental. Previous studies have shown ME/CFS-associated alterations in the immune system and mitochondria. We used transmission electron microscopy (TEM) to investigate the morphology and ultrastructure of unstimulated and stimulated ME/CFS immune cells and their intracellular organelles, including mitochondria. PBMCs from four participants were studied: a pair of identical twins discordant for moderate ME/CFS, as well as two age- and gender- matched unrelated subjects-one with an extremely severe form of ME/CFS and the other healthy. TEM analysis of CD3/CD28-stimulated T cells suggested a significant increase in the levels of apoptotic and necrotic cell death in T cells from ME/CFS patients (over 2-fold). Stimulated Tcells of ME/CFS patients also had higher numbers of swollen mitochondria. We also found a large increase in intracellular giant lipid droplet-like organelles in the stimulated PBMCs from the extremely severe ME/CFS patient potentially indicative of a lipid storage disorder. Lastly, we observed a slight increase in platelet aggregation in stimulated cells, suggestive of a possible role of platelet activity in ME/CFS pathophysiology and disease severity. These results indicate extensive morphological alterations in the cellular and mitochondrial phenotypes of ME/CFS patients' immune cells and suggest new insights into ME/CFS biology.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rajan D. Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, and VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - John J. Perrino
- Stanford Cell Sciences Imaging Facility (CSIF), Stanford University School of Medicine Stanford, Stanford, California, United States of America
| | - Damek V. Spacek
- Karius Incorporated, Redwood City, California, United States of America
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
12
|
Li MJ, Yan SB, Chen G, Li GS, Yang Y, Wei T, He DS, Yang Z, Cen GY, Wang J, Liu LY, Liang ZJ, Chen L, Yin BT, Xu RX, Huang ZG. Upregulation of CCNB2 and Its Perspective Mechanisms in Cerebral Ischemic Stroke and All Subtypes of Lung Cancer: A Comprehensive Study. Front Integr Neurosci 2022; 16:854540. [PMID: 35928585 PMCID: PMC9344069 DOI: 10.3389/fnint.2022.854540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclin B2 (CCNB2) belongs to type B cell cycle family protein, which is located on chromosome 15q22, and it binds to cyclin-dependent kinases (CDKs) to regulate their activities. In this study, 103 high-throughput datasets related to all subtypes of lung cancer (LC) and cerebral ischemic stroke (CIS) with the data of CCNB2 expression were collected. The analysis of standard mean deviation (SMD) and summary receiver operating characteristic (SROC) reflecting expression status demonstrated significant up-regulation of CCNB2 in LC and CIS (Lung adenocarcinoma: SMD = 1.40, 95%CI [0.98–1.83], SROC = 0.92, 95%CI [0.89–0.94]. Lung squamous cell carcinoma: SMD = 2.56, 95%CI [1.64–3.48]. SROC = 0.97, 95%CI [0.95–0.98]. Lung small cell carcinoma: SMD = 3.01, 95%CI [2.01–4.01]. SROC = 0.98, 95%CI [0.97–0.99]. CIS: SMD = 0.29, 95%CI [0.05–0.53], SROC = 0.68, 95%CI [0.63–0.71]). Simultaneously, protein-protein interaction (PPI) analysis indicated that CCNB2 is the hub molecule of crossed high-expressed genes in CIS and LC. Through Multiscale embedded gene co-expression network analysis (MEGENA), a gene module of CIS including 76 genes was obtained and function enrichment analysis of the CCNB2 module genes implied that CCNB2 may participate in the processes in the formation of CIS and tissue damage caused by CIS, such as “cell cycle,” “protein kinase activity,” and “glycosphingolipid biosynthesis.” Afterward, via single-cell RNA-seq analysis, CCNB2 was found up-regulated on GABAergic neurons in brain organoids as well as T cells expressing proliferative molecules in LUAD. Concurrently, the expression of CCNB2 distributed similarly to TOP2A as a module marker of cell proliferation in cell cluster. These findings can help in the field of the pathogenesis of LC-related CIS and neuron repair after CIS damage.
Collapse
Affiliation(s)
- Ming-Jie Li
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shi-Bai Yan
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guo-Sheng Li
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yue Yang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tao Wei
- Department of Neurology, Liuzhou People’s Hospital, Liuzhou, China
| | - De-Shen He
- The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou Gongren Hospital, Wuzhou, China
| | - Zhen Yang
- Department of Gerontology, No. 923 Hospital of Chinese People’s Liberation Army, Nanning, China
| | - Geng-Yu Cen
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun Wang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liu-Yu Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhi-Jian Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bin-Tong Yin
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruo-Xiang Xu
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhi-Guang Huang
- Department of Pathology/Forensic Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Zhi-Guang Huang,
| |
Collapse
|
13
|
Li M, Wang ZW, Fang LJ, Cheng SQ, Wang X, Liu NF. Programmed cell death in atherosclerosis and vascular calcification. Cell Death Dis 2022; 13:467. [PMID: 35585052 PMCID: PMC9117271 DOI: 10.1038/s41419-022-04923-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022]
Abstract
The concept of cell death has been expanded beyond apoptosis and necrosis to additional forms, including necroptosis, pyroptosis, autophagy, and ferroptosis. These cell death modalities play a critical role in all aspects of life, which are noteworthy for their diverse roles in diseases. Atherosclerosis (AS) and vascular calcification (VC) are major causes for the high morbidity and mortality of cardiovascular disease. Despite considerable advances in understanding the signaling pathways associated with AS and VC, the exact molecular basis remains obscure. In the article, we review the molecular mechanisms that mediate cell death and its implications for AS and VC. A better understanding of the mechanisms underlying cell death in AS and VC may drive the development of promising therapeutic strategies.
Collapse
Affiliation(s)
- Min Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zhen-Wei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Li-Juan Fang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shou-Quan Cheng
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
14
|
Leng Y, Zhang Y, Li X, Wang Z, Zhuang Q, Lu Y. Receptor Interacting Protein Kinases 1/3: The Potential Therapeutic Target for Cardiovascular Inflammatory Diseases. Front Pharmacol 2021; 12:762334. [PMID: 34867386 PMCID: PMC8637748 DOI: 10.3389/fphar.2021.762334] [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: 08/21/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
The receptor interacting protein kinases 1/3 (RIPK1/3) have emerged as the key mediators in cell death pathways and inflammatory signaling, whose ubiquitination, phosphorylation, and inhibition could regulate the necroptosis and apoptosis effectually. Recently, more and more studies show great interest in the mechanisms and the regulator of RIPK1/3-mediated inflammatory response and in the physiopathogenesis of cardiovascular diseases. The crosstalk of autophagy and necroptosis in cardiomyocyte death is a nonnegligible conversation of cell death. We elaborated on RIPK1/3-mediated necroptosis, pathways involved, the latest regulatory molecules and therapeutic targets in terms of ischemia reperfusion, myocardial remodeling, myocarditis, atherosclerosis, abdominal aortic aneurysm, and cardiovascular transplantation, etc.
Collapse
Affiliation(s)
- Yiming Leng
- Clinical Research Center of the 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Ying Zhang
- Transplantation Center of the 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zeyu Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quan Zhuang
- Transplantation Center of the 3rd Xiangya Hospital, Central South University, Changsha, China.,Research Center of National Health Ministry on Transplantation Medicine, Changsha, China
| | - Yao Lu
- Clinical Research Center of the 3rd Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Farahi L, Sinha SK, Lusis AJ. Roles of Macrophages in Atherogenesis. Front Pharmacol 2021; 12:785220. [PMID: 34899348 PMCID: PMC8660976 DOI: 10.3389/fphar.2021.785220] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that may ultimately lead to local proteolysis, plaque rupture, and thrombotic vascular disease, resulting in myocardial infarction, stroke, and sudden cardiac death. Circulating monocytes are recruited to the arterial wall in response to inflammatory insults and differentiate into macrophages which make a critical contribution to tissue damage, wound healing, and also regression of atherosclerotic lesions. Within plaques, macrophages take up aggregated lipoproteins which have entered the vessel wall to give rise to cholesterol-engorged foam cells. Also, the macrophage phenotype is influenced by various stimuli which affect their polarization, efferocytosis, proliferation, and apoptosis. The heterogeneity of macrophages in lesions has recently been addressed by single-cell sequencing techniques. This article reviews recent advances regarding the roles of macrophages in different stages of disease pathogenesis from initiation to advanced atherosclerosis. Macrophage-based therapies for atherosclerosis management are also described.
Collapse
Affiliation(s)
- Lia Farahi
- Monoclonal Antibody Research Center, Avicenna Research Institute, Tehran, Iran
| | - Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
16
|
Zhang Y, Li H, Huang Y, Chen H, Rao H, Yang G, Wan Q, Peng Z, Bertin J, Geddes B, Reilly M, Tran JL, Wang M. Stage-Dependent Impact of RIPK1 Inhibition on Atherogenesis: Dual Effects on Inflammation and Foam Cell Dynamics. Front Cardiovasc Med 2021; 8:715337. [PMID: 34760938 PMCID: PMC8572953 DOI: 10.3389/fcvm.2021.715337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/24/2021] [Indexed: 11/30/2022] Open
Abstract
Objective: Atherosclerosis is an arterial occlusive disease with hypercholesterolemia and hypertension as common risk factors. Advanced-stage stenotic plaque, which features inflammation and necrotic core formation, is the major reason for clinical intervention. Receptor interacting serine/threonine-protein kinase 1 (RIPK1) mediates inflammation and cell death and is expressed in atherosclerotic lesions. The role of RIPK1 in advanced-stage atherosclerosis is unknown. Approach and Results: To investigate the effect of RIPK1 inhibition in advanced atherosclerotic plaque formation, we used ApoESA/SA mice, which exhibit hypercholesterolemia, and develop angiotensin-II mediated hypertension upon administration of doxycycline in drinking water. These mice readily develop severe atherosclerosis, including that in coronary arteries. Eight-week-old ApoESA/SA mice were randomized to orally receive a highly selective RIPK1 inhibitor (RIPK1i, GSK547) mixed with a western diet, or control diet. RIPK1i administration reduced atherosclerotic plaque lesion area at 2 weeks of treatment, consistent with suppressed inflammation (MCP-1, IL-1β, TNF-α) and reduced monocyte infiltration. However, administration of RIPK1i unexpectedly exacerbated atherosclerosis at 4 weeks of treatment, concomitant with increased macrophages and lipid deposition in the plaques. Incubation of isolated macrophages with oxidized LDL resulted in foam cell formation in vitro. RIPK1i treatment promoted such foam cell formation while suppressing the death of these cells. Accordingly, RIPK1i upregulated the expression of lipid metabolism-related genes (Cd36, Ppara, Lxrα, Lxrb, Srebp1c) in macrophage foam cells with ABCA1/ABCG1 unaltered. Furthermore, RIPK1i treatment inhibited ApoA1 synthesis in the liver and reduced plasma HDL levels. Conclusion: RIPK1 modulates the development of atherosclerosis in a stage-dependent manner, implicating both pro-atherosclerotic (monocyte infiltration and inflammation) and anti-atherosclerotic effects (suppressing foam cell accumulation and promoting ApoA1 synthesis). It is critical to identify an optimal therapeutic duration for potential clinical use of RIPK1 inhibitor in atherosclerosis or other related disease indications.
Collapse
Affiliation(s)
- Yuze Zhang
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huihui Li
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yonghu Huang
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guoli Yang
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Wan
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zekun Peng
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - John Bertin
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA, United States
| | - Brad Geddes
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA, United States
| | - Michael Reilly
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA, United States
| | - Jean-Luc Tran
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA, United States
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Li D, Yang Y, Li D, Pan J, Chu C, Liu G. Organic Sonosensitizers for Sonodynamic Therapy: From Small Molecules and Nanoparticles toward Clinical Development. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101976. [PMID: 34350690 DOI: 10.1002/smll.202101976] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/17/2021] [Indexed: 06/13/2023]
Abstract
Sonodynamic therapy (SDT) is a novel noninvasive therapeutic modality that combines low-intensity ultrasound and sonosensitizers. Versus photo-mediated therapy, SDT has the advantages of deeper tissue penetration, high accuracy, and less side effects. Sonosensitizers are critical for therapeutic efficacy during SDT and organic sonosensitizers are important because of their clear structure, easy monitoring, evaluation of drug metabolism, and clinical transformation. Notably, nanotechnology can be used in the field of sonosensitizers and SDT to overcome the inherent obstacles and achieve sustainable innovation. This review introduces organic small molecule sonosensitizers, nano organic sonosensitizers, and their clinical translation by providing ideas and references for the design of sonosensitizers and SDT so as to promote its transformation to clinical applications in the future.
Collapse
Affiliation(s)
- Dong Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Yang
- Department of Cardiovascular, Xiang'an Hospital of Xiamen University, Xiamen, 361102, China
| | - Dengfeng Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jie Pan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, 361102, China
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Engineering Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
18
|
Zhou T, DeRoo E, Yang H, Stranz A, Wang Q, Ginnan R, Singer HA, Liu B. MLKL and CaMKII Are Involved in RIPK3-Mediated Smooth Muscle Cell Necroptosis. Cells 2021; 10:cells10092397. [PMID: 34572045 PMCID: PMC8471540 DOI: 10.3390/cells10092397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Receptor interacting protein kinase 3 (RIPK3)-mediated smooth muscle cell (SMC) necroptosis has been shown to contribute to the pathogenesis of abdominal aortic aneurysms (AAAs). However, the signaling steps downstream from RIPK3 during SMC necroptosis remain unknown. In this study, the roles of mixed lineage kinase domain-like pseudokinase (MLKL) and calcium/calmodulin-dependent protein kinase II (CaMKII) in SMC necroptosis were investigated. We found that both MLKL and CaMKII were phosphorylated in SMCs in a murine CaCl2-driven model of AAA and that Ripk3 deficiency reduced the phosphorylation of MLKL and CaMKII. In vitro, mouse aortic SMCs were treated with tumor necrosis factor α (TNFα) plus Z-VAD-FMK (zVAD) to induce necroptosis. Our data showed that both MLKL and CaMKII were phosphorylated after TNFα plus zVAD treatment in a time-dependent manner. SiRNA silencing of Mlkl-diminished cell death and administration of the CaMKII inhibitor myristoylated autocamtide-2-related inhibitory peptide (Myr-AIP) or siRNAs against Camk2d partially inhibited necroptosis. Moreover, knocking down Mlkl decreased CaMKII phosphorylation, but silencing Camk2d did not affect phosphorylation, oligomerization, or trafficking of MLKL. Together, our results indicate that both MLKL and CaMKII are involved in RIPK3-mediated SMC necroptosis, and that MLKL is likely upstream of CaMKII in this process.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
| | - Elise DeRoo
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
| | - Huan Yang
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
| | - Amelia Stranz
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
| | - Qiwei Wang
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
| | - Roman Ginnan
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.G.); (H.A.S.)
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.G.); (H.A.S.)
| | - Bo Liu
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (T.Z.); (E.D.); (H.Y.); (A.S.); (Q.W.)
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
19
|
Wu Y, Zheng Z, Cao X, Yang Q, Norton V, Adini A, Maiti AK, Adini I, Wu H. RIP1/RIP3/MLKL Mediates Myocardial Function Through Necroptosis in Experimental Autoimmune Myocarditis. Front Cardiovasc Med 2021; 8:696362. [PMID: 34497836 PMCID: PMC8419468 DOI: 10.3389/fcvm.2021.696362] [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: 04/16/2021] [Accepted: 07/22/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiomyopathy often leads to dilated cardiomyopathy (DCM) when caused by viral myocarditis. Apoptosis is long considered as the principal process of cell death in cardiomyocytes, but programmed necrosis or necroptosis is recently believed to play an important role in cardiomyocyte cell death. We investigated the role of necroptosis and its interdependency with other processes of cell death, autophagy, and apoptosis in a rat system of experimental autoimmune myocarditis (EAM). We successfully created a rat model system of EAM by injecting porcine cardiac myosin (PCM) and showed that in EAM, all three forms of cell death increase considerably, resulting in the deterioration of cardiac conditions with an increase in inflammatory infiltration in cardiomyocytes. To explore whether necroptosis occurs in EAM rats independent of autophagy, we treated EAM rats with a RIP1/RIP3/MLKL kinase-mediated necroptosis inhibitor, Necrostatin-1 (Nec-1). In Nec-1 treated rats, cell death proceeds through apoptosis but has no significant effect on autophagy. In contrast, autophagy inhibitor 3-Methyl Adenine (3-MA) increases necroptosis, implying that blockage of autophagy must be compensated through necroptosis. Caspase 8 inhibitor zVAD-fmk blocks apoptosis but increases both necroptosis and autophagy. However, all necroptosis, apoptosis, and autophagy inhibitors independently reduce inflammatory infiltration in cardiomyocytes and improve cardiac conditions. Since apoptosis or autophagy is involved in many important cellular aspects, instead of suppressing these two major cell death processes, Nec1 can be developed as a potential therapeutic target for inflammatory myocarditis.
Collapse
Affiliation(s)
- Yujing Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China.,Department of Emergency, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhenzhong Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China
| | - Xiantong Cao
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qing Yang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Cardiology, Jiangxi Hypertension Research Institute, Nanchang, China
| | - Vikram Norton
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Avner Adini
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Amit K Maiti
- Mydnavar, Department of Genetics and Genomics, Troy, MI, United States
| | - Irit Adini
- Center for Engineering in Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
20
|
Chen X, Deng Z, Feng J, Chang Q, Lu F, Yuan Y. Necroptosis in Macrophage Foam Cells Promotes Fat Graft Fibrosis in Mice. Front Cell Dev Biol 2021; 9:651360. [PMID: 33842478 PMCID: PMC8027326 DOI: 10.3389/fcell.2021.651360] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Fibrosis is a major grafting-related complication that leads to fat tissue dysfunction. Macrophage-induced inflammation is related to the development of fat tissue fibrosis. Necroptosis is a recently discovered pathway of programmed cell necrosis that results in severe inflammation and subsequent tissue fibrosis. Thus, in this study, we investigated the role of macrophage necroptosis in fat graft fibrosis and the underlying mechanisms. Methods: Fibrosis and necroptosis were investigated in mouse fat tissue before and after grafting. An in vitro “crown-like” structure (CLS) cell culture model was developed by co-culturing RAW 264.7 macrophages with apoptotic adipocytes to reproduce in vivo CLS macrophage-adipocyte interactions. Lipid uptake and necroptosis in CLS macrophages were analyzed using Oil-Red-O staining, western blotting, and immunofluorescence. RAW264.7 macrophages were cultured alone or with apoptotic adipocytes and treated with a necroptosis inhibitor (Nec-1 or GSK872) to explore the paracrine effect of necroptotic CLS macrophages on collagen synthesis in fibroblasts in vitro. Mice were treated with Nec-1 to analyze the effect of blocking necroptosis on fat graft fibrosis. Results: Fibrosis was increased after grafting in fat grafts of mice. Macrophages clustered around apoptotic adipocytes or large oil droplets to form a typical CLS in fibrotic depots. This was accompanied by formation and necroptosis of macrophage foam cells (MFCs) in CLSs. RAW 264.7 macrophages co-cultured with apoptotic adipocytes induced CLS formation in vitro, and lipid accumulation in CLS macrophages resulted in the formation and necroptosis of MFCs. Necroptosis of MFCs altered the expression of collagen I and VI in fibroblasts via a paracrine mechanism involving inflammatory cytokines/chemokines, which was reversed by GSK872 or Nec-1 treatment. Furthermore, treatment with Nec-1 ameliorated fat graft fibrosis in mice. Conclusion: Apoptotic adipocytes induced necroptosis of MFCs, and necroptosis of these cells activated collagen synthesis in fibroblasts via a paracrine mechanism. Inhibition of necroptosis in macrophages is a potential approach to prevent fibrosis in fat grafts.
Collapse
Affiliation(s)
- Xihang Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zilong Deng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingwei Feng
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Yuan
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Impact of myeloid RIPK1 gene deletion on atherogenesis in ApoE-deficient mice. Atherosclerosis 2021; 322:51-60. [PMID: 33706083 DOI: 10.1016/j.atherosclerosis.2021.02.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/23/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Targeting macrophage death is a promising strategy for stabilizing atherosclerotic plaques. Recently, necroptosis was identified as a form of regulated necrosis in atherosclerosis. Receptor-interacting serine/threonine-protein kinase (RIPK)1 is an upstream regulator of RIPK3, which is a crucial kinase for necroptosis induction. We aimed to investigate the impact of myeloid-specific RIPK1 gene deletion on atherogenesis. METHODS RIPK1F/FLysM-Cre+ApoE-/- and RIPK1+/+LysM-Cre+ApoE-/- mice were fed a western-type diet (WD) for 16 or 24 weeks to induce plaque formation. RESULTS After 16 weeks WD, plaque area and percentage necrosis in RIPK1F/FLysM-Cre+ApoE-/- mice were significantly decreased as compared to plaques of RIPK1+/+LysM-Cre+ApoE-/- mice. Moreover, plaques of RIPK1F/FLysM-Cre+ApoE-/- mice showed more apoptosis and a decreased macrophage content. After 24 weeks WD, plaque size and percentage necrosis were no longer different between the two groups. Free apoptotic cells strongly accumulated in plaques of RIPK1F/FLysM-Cre+ApoE-/- mice. In addition to apoptosis, necroptosis was upregulated in plaques of RIPK1F/FLysM-Cre+ApoE-/- mice. In vitro, TNF-α triggered apoptosis in RIPK1F/FLysM-Cre+ApoE-/-, but not in RIPK1+/+LysM-Cre+ApoE-/- macrophages. Moreover, RIPK1F/FLysM-Cre+ApoE-/- macrophages were not protected against RIPK3-dependent necroptosis. CONCLUSIONS The impact of myeloid RIPK1 gene deletion depends on the stage of atherogenesis. At 16 weeks WD, myeloid RIPK1 gene deletion resulted in increased apoptosis, thereby slowing down plaque progression. However, despite decreased macrophage content, plaque and necrotic core size were no longer reduced after 24 weeks of WD, most likely due to the accumulation of free apoptotic and necroptotic cells.
Collapse
|
22
|
Lu LQ, Tian J, Luo XJ, Peng J. Targeting the pathways of regulated necrosis: a potential strategy for alleviation of cardio-cerebrovascular injury. Cell Mol Life Sci 2021; 78:63-78. [PMID: 32596778 PMCID: PMC11072340 DOI: 10.1007/s00018-020-03587-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Apoptosis, necrosis and autophagy-dependent cell death are the three major types of cell death. Traditionally, necrosis is thought as a passive and unregulated form of cell death. However, certain necrosis can also occur in a highly regulated manner, referring to regulated necrosis. Depending on the signaling pathways, regulated necrosis can be further classified as necroptosis, pyroptosis, ferroptosis, parthanatos and CypD-mediated necrosis. Numerous studies have reported that regulated necrosis contributes to the progression of multiple injury-relevant diseases. For example, necroptosis contributes to the development of myocardial infarction, atherosclerosis, heart failure and stroke; pyroptosis is involved in the progression of myocardial or cerebral infarction, atherosclerosis and diabetic cardiomyopathy; while ferroptosis, parthanatos and CypD-mediated necrosis participate in the pathological process of myocardial and/or cerebral ischemia/reperfusion injury. Thereby, targeting the pathways of regulated necrosis pharmacologically or genetically could be an efficient strategy for reducing cardio-cerebrovascular injury. Further study needs to focus on the crosstalk and interplay among different types of regulated necrosis. Pharmacological intervention of two or more types of regulated necrosis simultaneously may have advantages in clinic to treat injury-relevant diseases.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jing Tian
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
23
|
Li L, Tan H, Zou Z, Gong J, Zhou J, Peng N, Su L, Maegele M, Cai D, Gu Z. Preventing necroptosis by scavenging ROS production alleviates heat stress-induced intestinal injury. Int J Hyperthermia 2020; 37:517-530. [PMID: 32423248 DOI: 10.1080/02656736.2020.1763483] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Worldwide heat stroke incidence has increased in recent years and is associated with high morbidity and mortality. Therefore, it is critical to identify mechanisms that mediate heat stroke. Previous studies suggested that damage to the small intestine may be a major factor in heat stroke-related morbidity and mortality. However, the mechanism underlying heat stroke related small intestine injury remains unclear.Methods: To explore how heat stroke promotes intestinal damage, we applied two well established models: mouse and IEC-6 cells heat stress (HS) to mimic heat stroke both in vivo and in vitro. The percentages of viability and cell death were assessed by WST-1 and LDH release assays. Induction of HS-induced cell death was analyzed by flow cytometry with Annexin V-FITC/PI staining. Flow cytometry was used to analyze HS-induced mitochondrial superoxide with MitoSOX staining. Malondialdehyde (MDA) levels and superoxide dismutase (SOD) levels were detected by ELISA. Flow cytometry was used to analyze HS-induced mitochondrial depolarization (low ΔΨm) with JC-1 staining. Histopathology changes in the ileum were detected by H&E staining.The ileum ultrastructure was observed by transmission electron microscopy (TEM). RIPK1, RIPK3, phosphorylated MLKL, and MLKL levels were detected by Western blot. RIPK1-RIPK3 complexes were measured by immunoprecipitation assay.Results: HS increased both necrotic cell rate and RIPK1, RIPK3, and phosphorylated MLKL expression levels in IEC-6 cells. These increased expression levels promoted higher RIPK1-RIPK3 complex formation, leading to necrosome formation both in vivo and in vitro. Moreover, HS caused dyshomeostasis, an oxidative stress response, and mitochondrial damage, along with small intestinal tissue injury and cell death. However, IEC-6 cells or mice pretreated with the RIPK1 activity chemical inhibitor Nec-1 or RIPK3 activity chemical inhibitor GSK'872 significantly reversed these phenomena and promoted balance in oxidative stress response homeostasis. More importantly, the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) pretreatment significantly inhibited HS-induced RIPK1/RIPK3-dependent necroptosis formation both in vivo and in vitro, suggesting that preventing necroptosis via scavenging ROS production might alleviate HS-induced small intestinal tissue injury and cell death.Conclusion: This study provides strong evidence that HS causes damage to both the small intestine and intestinal epithelial cells, scavenging ROS production can significantly alleviate such RIPK1/RIPK3-dependent necroptosis, mediating HS-induced intestinal damage both in vitro and in vivo. These findings provide a clear target for future mechanism-based therapeutic strategies for patients diagnosed with heat stroke.
Collapse
Affiliation(s)
- Li Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China
| | - Hongping Tan
- Department of epilepsy centre, Guangdong Sanjiu Brain Hospital, Guangzhou China
| | - Zhimin Zou
- Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China
| | - Jian Gong
- Department of Intensive Care Unit, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Junjie Zhou
- Department of Intensive Care Unit, Heyuan People's Hospital, Heyuan, Guangdong, China
| | - Na Peng
- Department of Intensive Care Unit, General Hospital of Southern Theater Command, PLA; Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, Guangzhou, Guangdong, China
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Southern Theater Command, PLA; Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, Guangzhou, Guangdong, China
| | - Marc Maegele
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Treatment Center for Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center (CMMC), University Witten/Herdecke (UW/H), Cologne, Germany
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengtao Gu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China.,Department of Treatment Center for Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Cao L, Mu W. Necrostatin-1 and necroptosis inhibition: Pathophysiology and therapeutic implications. Pharmacol Res 2020; 163:105297. [PMID: 33181319 PMCID: PMC7962892 DOI: 10.1016/j.phrs.2020.105297] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/17/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Necrostatin-1 (Nec-1) is a RIP1-targeted inhibitor of necroptosis, a form of programmed cell death discovered and investigated in recent years. There are already many studies demonstrating the essential role of necroptosis in various diseases, including inflammatory diseases, cardiovascular diseases and neurological diseases. However, the potential of Nec-1 in diseases has not received much attention. Nec-1 is able to inhibit necroptosis signaling pathway and thus ameliorate necroptotic cell death in disease development. Recent research findings indicate that Nec-1 could be applied in several types of diseases to alleviate disease development or improve prognosis. Moreover, we predict that Nec-1 has the potential to protect against the complications of coronavirus disease 2019 (COVID-19). This review summarized the effect of Nec-1 in disease models and the underlying molecular mechanism, providing research evidence for its future application.
Collapse
Affiliation(s)
- Liyuan Cao
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Mu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
25
|
DeRoo E, Zhou T, Liu B. The Role of RIPK1 and RIPK3 in Cardiovascular Disease. Int J Mol Sci 2020; 21:E8174. [PMID: 33142926 PMCID: PMC7663726 DOI: 10.3390/ijms21218174] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases, including peripheral arterial and venous disease, myocardial infarction, and stroke, are the number one cause of death worldwide annually. In the last 20 years, the role of necroptosis, a newly identified form of regulated necrotic cell death, in cardiovascular disease has come to light. Specifically, the damaging role of two kinase proteins pivotal in the necroptosis pathway, Receptor Interacting Protein Kinase 1 (RIPK1) and Receptor Interacting Protein Kinase 3 (RIPK3), in cardiovascular disease has become a subject of great interest and importance. In this review, we provide an overview of the current evidence supporting a pathologic role of RIPK1 and RIPK3 in cardiovascular disease. Moreover, we highlight the evidence behind the efficacy of targeted RIPK1 and RIPK3 inhibitors in the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Bo Liu
- Department of Surgery, Division of Vascular Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (E.D.); (T.Z.)
| |
Collapse
|
26
|
Early modulation of macrophage ROS-PPARγ-NF-κB signalling by sonodynamic therapy attenuates neointimal hyperplasia in rabbits. Sci Rep 2020; 10:11638. [PMID: 32669704 PMCID: PMC7363872 DOI: 10.1038/s41598-020-68543-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/14/2020] [Indexed: 11/08/2022] Open
Abstract
Disruption of re-endothelialization and haemodynamic balance remains a critical side effect of drug-eluting stents (DES) for preventing intimal hyperplasia. Previously, we found that 5-aminolevulinic acid-mediated sonodynamic therapy (ALA-SDT) suppressed macrophage-mediated inflammation in atherosclerotic plaques. However, the effects on intimal hyperplasia and re-endothelialization remain unknown. In this study, 56 rabbits were randomly assigned to control, ultrasound, ALA and ALA-SDT groups, and each group was divided into two subgroups (n = 7) on day 3 after right femoral artery balloon denudation combined with a hypercholesterolemic diet. Histopathological analysis revealed that ALA-SDT enhanced macrophage apoptosis and ameliorated inflammation from day 1. ALA-SDT inhibited neointima formation without affecting re-endothelialization, increased blood perfusion, decreased the content of macrophages, proliferating smooth muscle cells (SMCs) and collagen but increased elastin by day 28. In vitro, ALA-SDT induced macrophage apoptosis and reduced TNF-α, IL-6 and IL-1β via the ROS-PPARγ-NF-κB signalling pathway, which indirectly inhibited human umbilical artery smooth muscle cell (HUASMC) proliferation, migration and IL-6 production. ALA-SDT effectively inhibits intimal hyperplasia without affecting re-endothelialization. Hence, its clinical application combined with bare-metal stent (BMS) implantation presents a potential strategy to decrease bleeding risk caused by prolonged dual-antiplatelet regimen after DES deployment.
Collapse
|
27
|
Yang B, Qin Q, Xu L, Lv X, Liu Z, Song E, Song Y. Polychlorinated Biphenyl Quinone Promotes Atherosclerosis through Lipid Accumulation and Endoplasmic Reticulum Stress via CD36. Chem Res Toxicol 2020; 33:1497-1507. [PMID: 32434321 DOI: 10.1021/acs.chemrestox.0c00123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic environmental pollutants. According to previous epidemiological reports, PCBs exposure is highly related to atherosclerosis. However, studies of PCBs metabolites and atherosclerosis and corresponding mechanism studies are scarce. In this study, we evaluated the effect of 2,3,5-trichloro-6-phenyl-[1,4]-benzoquinone (PCB29-pQ), a presumptive PCB metabolite, on atherosclerosis. Aortic plaques were increased in PCB29-pQ-treated ApoE-/- mice [intraperitoneally (i.p.) injection of 5 mg/kg body weight of PCB29-pQ once a week for 12 continuous weeks, high-fat feeding]. We observed lipids accumulation and the release of interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) in ApoE-/- mice. In addition, we found that PCB29-pQ promoted the levels of total cholesterol, free cholesterol, triglyceride, and cholesteryl ester. Mechanism investigation indicated that PCB29-pQ induces the activation of three branches of endoplasmic reticulum (ER) stress response, that is, phosphorylated protein kinase R-like ER kinase (p-PERK), eukaryotic translation initiation factor 2α (eIF2α) and transcription factor 6 (ATF6), which is responsible for downstream necrosis. More importantly, we found the silence of CD36 is able to reverse PCB29-pQ-induced adverse effects completely. Overall, PCB29-pQ exposure resulted in lipid accumulation, ER stress response, apoptosis, and pro-inflammatory cytokines release via CD36, ultimately leading to atherosclerosis.
Collapse
Affiliation(s)
- Bingwei Yang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Qi Qin
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Lei Xu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Xuying Lv
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Zixuan Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Erqun Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Yang Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
28
|
Caspase-8 Regulates Endoplasmic Reticulum Stress-Induced Necroptosis Independent of the Apoptosis Pathway in Auditory Cells. Int J Mol Sci 2019; 20:ijms20235896. [PMID: 31771290 PMCID: PMC6928907 DOI: 10.3390/ijms20235896] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/15/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
The aim of this study is to elucidate the detailed mechanism of endoplasmic reticulum (ER) stress-induced auditory cell death based on the function of the initiator caspases and molecular complex of necroptosis. Here, we demonstrated that ER stress initiates not only caspase-9-dependent intrinsic apoptosis along with caspase-3, but also receptor-interacting serine/threonine kinase (RIPK)1-dependent necroptosis in auditory cells. We observed the ultrastructural characteristics of both apoptosis and necroptosis in tunicamycin-treated cells under transmission electron microscopy (TEM). We demonstrated that ER stress-induced necroptosis was dependent on the induction of RIPK1, negatively regulated by caspase-8 in auditory cells. Our data suggested that ER stress-induced intrinsic apoptosis depends on the induction of caspase-9 along with caspase-3 in auditory cells. The results of this study reveal that necroptosis could exist for the alternative backup cell death route of apoptosis in auditory cells under ER stress. Interestingly, our data results in a surge in the recognition that therapies aimed at the inner ear protection effect by caspase inhibitors like zVAD-fmk might arrest apoptosis but can also have the unanticipated effect of promoting necroptosis. Thus, RIPK1-dependent necroptosis would be a new therapeutic target for the treatment of sensorineural hearing loss due to ER stress.
Collapse
|
29
|
Yang B, Wang Y, Qin Q, Xia X, Liu Z, Song E, Song Y. Polychlorinated Biphenyl Quinone Promotes Macrophage-Derived Foam Cell Formation. Chem Res Toxicol 2019; 32:2422-2432. [PMID: 31680514 DOI: 10.1021/acs.chemrestox.9b00184] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Polychlorinated biphenyls (PCBs) are organic environmental pollutants that are accused of various toxic effects. PCB exposure is widely believed to be associated with atherosclerosis, but the underlying mechanisms are unclear. Although PCBs are easily metabolized, there is rarely information on the effects of their metabolites on atherosclerosis. Currently, we evaluate the effect of 2,3,5-trichloro-6-phenyl-[1,4]-benzoquinone (PCB29-pQ) on the critical phase of atherosclerosis development, that is, the formation of macrophage-derived foam cells. We exposed Ox-LDL-induced RAW264.7 cells to 2.5 μM and 5 μM PCB29-pQ. Varieties of evidence have demonstrated that PCB29-pQ promotes foam cell formation and develops proinflammatory cascade and cell necroptosis. In detail, we observed that PCB29-pQ increased levels of total cholesterol (TC), free cholesterol (FC), triglyceride (TG), and cholesteryl ester (CE) by increasing the cholesterol influx and reducing the cholesterol efflux. Moreover, we found that PCB29-pQ induced inflammatory cytokines, such as tumor necrosis factor (TNF-α), interleukin 6 (IL-6), and IL-1β, released by activating the mitogen-activated protein kinase (MAPK)-nuclear factor kappa B (NF-κB) inflammatory pathway. In addition, we demonstrated that PCB29-pQ induced cell necroptosis via receptor interacting protein kinases 1 and 3 (RIPK1/3) and a mixed-lineage kinase domain-like (MLKL) pathway. Finally, the overproduction of reactive oxygen species (ROS) by PCB29-pQ played significant roles in these processes, which could be reversed with an antioxidant. Overall, our results indicated that PCB29-pQ promoted the macrophage formation of foam cells, inflammation, and cell necroptosis.
Collapse
Affiliation(s)
- Bingwei Yang
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Yawen Wang
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Qi Qin
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Xiaomin Xia
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Zixuan Liu
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Erqun Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| | - Yang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences , Southwest University , Chongqing , People's Republic of China , 400715
| |
Collapse
|
30
|
Implications of Necroptosis for Cardiovascular Diseases. Curr Med Sci 2019; 39:513-522. [PMID: 31346984 DOI: 10.1007/s11596-019-2067-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 03/01/2019] [Indexed: 02/08/2023]
Abstract
Necroptosis is a non-apoptotic programmed cell death pathway, which causes necrosis-like morphologic changes and triggers inflammation in the surrounding tissues. Accumulating evidence has demonstrated that necroptosis is involved in a number of pathological processes that lead to cardiovascular diseases. However, the exact molecular pathways linking them remain unknown. Herein, this review summarizes the necroptosis-related pathways involved in the development of various cardiovascular diseases, including atherosclerosis, cardiac ischemia-reperfusion injury, cardiac hypertrophy, dilated cardiomyopathy and myocardial infarction, and may shed light on the diagnosis and treatment of these diseases.
Collapse
|
31
|
Guo T, Liu T, Sun Y, Liu X, Xiong R, Li H, Li Z, Zhang Z, Tian Z, Tian Y. Sonodynamic therapy inhibits palmitate-induced beta cell dysfunction via PINK1/Parkin-dependent mitophagy. Cell Death Dis 2019; 10:457. [PMID: 31186419 PMCID: PMC6560035 DOI: 10.1038/s41419-019-1695-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022]
Abstract
In type 2 diabetes mellitus (T2DM), the overload of glucose and lipids can promote oxidative stress and inflammatory responses and contribute to the failure of beta cells. However, therapies that can modulate the function of beta cells and thus prevent their failure have not been well explored. In this study, beta cell injury model was established with palmitic acid (PA) to simulate the lipotoxicity (high-fat diet) found in T2DM. Sonodynamic therapy (SDT), a novel physicochemical treatment, was applied to treat injured beta cells. We found that SDT had specific effects on mitochondria and induced transient large amount of mitochondrial reactive oxygen species (ROS) production in beta cells. SDT also improved the morphology and function of abnormal mitochondria, inhibited inflammatory response and reduced beta cell dysfunction. The improvement of mitochondria was mediated by PINK1/Parkin-dependent mitophagy. Additionally, SDT rescued the transcription of PINK1 mRNA which was blocked by PA treatment, thus providing abundant PINK1 for mitophagy. Moreover, SDT also increased insulin secretion from beta cells. The protective effects of SDT were abrogated when mitophagy was inhibited by cyclosporin A (CsA). In summary, SDT potently inhibits lipotoxicity-induced beta cell failure via PINK1/Parkin-dependent mitophagy, providing theoretical guidance for T2DM treatment in aspects of islet protection.
Collapse
Affiliation(s)
- Tian Guo
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Tianyang Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Yun Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Xianna Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Rongguo Xiong
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - He Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Zhitao Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Zhiguo Zhang
- Laboratory of Photo- and Sono-theranostic Technologies and Condensed Matter Science and Technology Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhen Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China. .,Key Laboratory of Acoustic Photoelectric Magnetic Diagnosis and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, 150081, China.
| | - Ye Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China. .,Key Laboratory of Acoustic Photoelectric Magnetic Diagnosis and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, 150081, China. .,Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
32
|
Martinet W, Coornaert I, Puylaert P, De Meyer GRY. Macrophage Death as a Pharmacological Target in Atherosclerosis. Front Pharmacol 2019; 10:306. [PMID: 31019462 PMCID: PMC6458279 DOI: 10.3389/fphar.2019.00306] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual build-up of plaques within the vessel wall of middle-sized and large arteries. Over the past decades, treatment of atherosclerosis mainly focused on lowering lipid levels, which can be accomplished by the use of statins. However, some patients do not respond sufficiently to statin therapy and therefore still have a residual cardiovascular risk. This issue highlights the need for novel therapeutic strategies. As macrophages are implicated in all stages of atherosclerotic lesion development, they represent an important alternative drug target. A variety of anti-inflammatory strategies have recently emerged to treat or prevent atherosclerosis. Here, we review the canonical mechanisms of macrophage death and their impact on atherogenesis and plaque stability. Macrophage death is a prominent feature of advanced plaques and is a major contributor to necrotic core formation and plaque destabilization. Mechanisms of macrophage death in atherosclerosis include apoptosis, passive or accidental necrosis as well as secondary necrosis, a type of death that typically occurs when apoptotic cells are insufficiently cleared by neighboring cells via a phagocytic process termed efferocytosis. In addition, less-well characterized types of regulated necrosis in macrophages such as necroptosis, pyroptosis, ferroptosis, and parthanatos may occur in advanced plaques and are also discussed. Autophagy in plaque macrophages is an important survival pathway that protects against cell death, yet massive stimulation of autophagy promotes another type of death, usually referred to as autosis. Multiple lines of evidence indicate that a better insight into the different mechanisms of macrophage death, and how they mutually interact, will provide novel pharmacological strategies to resolve atherosclerosis and stabilize vulnerable, rupture-prone plaques.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Isabelle Coornaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
33
|
Huang L, Chen Q, Yu L, Bai D. Pyropheophorbide-α methyl ester-mediated photodynamic therapy induces apoptosis and inhibits LPS-induced inflammation in RAW264.7 macrophages. Photodiagnosis Photodyn Ther 2018; 25:148-156. [PMID: 30562579 DOI: 10.1016/j.pdpdt.2018.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/05/2018] [Accepted: 12/07/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND This study aimed to determine the effect of pyropheophorbide-α methyl ester (MPPa)-mediated photodynamic therapy (MPPa-PDT) on the apoptosis and inflammation of murine macrophage RAW264.7 cells. METHODS Uptake and subcellular localization of MPPa was detected by flow cytometry and confocal fluorescence microscope. Cell viability was assessed by CCK-8; ROS levels were assessed by DCFH-DA. Cell apoptosis was measured by flow cytometry and Hoechst 33342 staining, whereas mitochondrial membrane potential was detected by JC-1 staining. Secretion of tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) was determined using ELISA kits. Caspase-3, cleaved caspase-3, procaspase-9, cleaved caspase-9, PARP, cleaved PARP, Bcl-2, Bax, NF-κB p-p65, p-IKKα/β, and p-IκBα were measured by western blotting. Nuclear factor κB (NF-κB)-p65 nuclear translocation was observed by immunofluorescence. RESULTS MPPa -PDT influenced cell viability in a light dose-dependent manner. It induced ROS formation and RAW264.7 cell apoptosis. It also increased the expression of cleaved caspase-3, cleaved caspase-9, cleaved PARP and Bax, decreased the expression of Bcl-2. While TNF-α, IL-1β, and IL-6 increased in LPS group (model of inflammation), it deceased in LPS-MPPa-PDT group. NF-κB p-p65, p-IKKα/β, and p-IκBα had higher expression in LPS group while that reduced in LPS-MPPa-PDT group. Simultaneously, MPPa-PDT inhibited nuclear translocation of NF-κB-p65 caused by LPS. CONCLUSIONS MPPa-PDT can induce apoptosis and attenuate inflammation in mouse RAW264.7 macrophages, thereby suggesting a promising therapy for atherosclerosis.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Qing Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Lehua Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
34
|
Jing L, Song F, Liu Z, Li J, Wu B, Fu Z, Jiang J, Chen Z. MLKL-PITPα signaling-mediated necroptosis contributes to cisplatin-triggered cell death in lung cancer A549 cells. Cancer Lett 2018; 414:136-146. [PMID: 29104146 DOI: 10.1016/j.canlet.2017.10.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/28/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022]
Abstract
Necroptosis has been reported to be involved in cisplatin-induced cell death, but the mechanisms underlying the occurrence of necroptosis are not fully elucidated. In this study, we show that apart from apoptosis, cisplatin induces necroptosis in A549 cells. The alleviation of cell death by two necroptosis inhibitors-necrostatin-1 (Nec-1) and necrosulfonamide (NSA), and the phosphorylation of mixed lineage kinase domain-like protein (MLKL) at serine 358, suggest the involvement of receptor-interacting protein kinase 1 (RIPK1)-RIPK3-MLKL signaling in cisplatin-treated A549 cells. Additionally, the initiation of cisplatin-induced necroptosis relies on autocrine tumor necrosis factor alpha (TNF-α). Furthermore, we present the first evidence that phosphatidylinositol transfer protein alpha (PITPα) is involved in MLKL-mediated necroptosis by interacting with the N terminal MLKL on its sixth helix and the preceding loop, which facilitates MLKL oligomerization and plasma membrane translocation in necroptosis. Silencing of PITPα expression interferes with MLKL function and reduces cell death. Our data elucidate that cisplatin-treated lung cancer cells undergo a new type of programmed cell death called necroptosis and shed new light on how MLKL translocates to the plasma membrane.
Collapse
Affiliation(s)
- Lin Jing
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Fei Song
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Zhenyu Liu
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Jianghua Li
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Bo Wu
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Zhiguang Fu
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Jianli Jiang
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China.
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine, Cell Engineering Research Center and Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China.
| |
Collapse
|
35
|
Gonçalvez KDO, Vieira DP, Courrol LC. Study of THP-1 Macrophage Viability after Sonodynamic Therapy Using Methyl Ester of 5-Aminolevulinic Acid Gold Nanoparticles. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:2009-2017. [PMID: 29936026 DOI: 10.1016/j.ultrasmedbio.2018.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/03/2018] [Accepted: 05/11/2018] [Indexed: 06/08/2023]
Abstract
Sonodynamic therapy (SDT) is emerging as new atherosclerosis treatment. The use of gold nanoparticles (AuNPs) as the vehicle for a sensitizer delivery improves reactive oxygen species formation. In this study, methyl ester of aminolevulinic acid (MALA) gold nanoparticles (MALA:AuNPs) functionalized with polyethylene glycol (PEG) were synthesized by photoreduction and characterized by ultraviolet/visible optical absorption, zeta potential and electron microscopy. The reactive oxygen species generation induced by ultrasound irradiation of MALA:AuNPs solutions was studied by observing the decrease in the 1,3-diphenylisobenzofuran emission band. The potential use of MALA:AuNPs as sensitizer for sonodynamic therapy was investigated on THP-1 macrophages. The cytotoxicity test was also described. The findings suggested that ultrasound combined with MALA:AuNPs provides impressive results in in vitro studies. Sonodynamic therapy with MALA:AuNPs through 2 minutes of ultrasound exposure (1 MHz and 1 W/cm2) culminated with total macrophage reduction. Thus, sonodynamic therapy combined with MALA:AuNPs has potential as a treatment for atherosclerosis.
Collapse
Affiliation(s)
- Karina de Oliveira Gonçalvez
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Física, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | - Daniel Perez Vieira
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares, São Paulo, São Paulo, Brazil
| | - Lilia Coronato Courrol
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Física, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil.
| |
Collapse
|
36
|
Zhe-Wei S, Li-Sha G, Yue-Chun L. The Role of Necroptosis in Cardiovascular Disease. Front Pharmacol 2018; 9:721. [PMID: 30034339 PMCID: PMC6043645 DOI: 10.3389/fphar.2018.00721] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 06/13/2018] [Indexed: 01/14/2023] Open
Abstract
A newly discovered mechanism of cell death, programmed necrosis (necroptosis), combines features of both necrosis and apoptosis. Necroptosis is tightly modulated by a series of characteristic signaling pathways. Activating necroptosis by ligands of death receptors requires the kinase activity of receptor-interacting protein 1 (RIP1), which mediates the activation of receptor-interacting protein 3 (RIP3) and mixed lineage kinase domain-like (MLKL) two critical downstream mediators of necroptosis. Recently, different cytokines have been found participating in this mechanism of cell death. Necroptosis has been proposed as an important component to the pathophysiology of heart disease such as vascular atherosclerosis, ischemia-reperfusion injury, myocardial infarction and cardiac remodeling. Targeting necroptosis signaling pathways may provide therapeutic benefit in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Shi Zhe-Wei
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ge Li-Sha
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Yue-Chun
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
37
|
Qian J, Gao Q. Sonodynamic Therapy Mediated by Emodin Induces the Oxidation of Microtubules to Facilitate the Sonodynamic Effect. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:853-860. [PMID: 29398130 DOI: 10.1016/j.ultrasmedbio.2017.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 12/17/2017] [Accepted: 12/19/2017] [Indexed: 06/07/2023]
Abstract
In previous studies, sonodynamic therapy mediated by emodin (emodin-SDT) induced cytoskeletal filament disruption and apoptosis of THP-1-derived macrophages. In this research, we investigated the underlying mechanism. THP-1-derived macrophages were incubated with emodin and exposed to ultrasound irradiation. After emodin-SDT, we measured the production of reactive oxygen species (ROS) and analyzed the level of amino acid oxidation in microtubules, the cleavage of microtubules and the mitochondrial membrane potential (MMP). We found that intracellular emodin accumulated mainly on microtubules. After emodin-SDT, generation of ROS was evident. Analysis of the carbonyl content of proteins suggested oxidation of microtubules. Microtubules were disrupted after emodin-SDT, and the antioxidant N-acetyl-L-cysteine prevented this disruption. MMP decreased after emodin-SDT, and this effect could be prevented by N-acetyl-L-cysteine. We conclude that emodin-SDT induces the generation of ROS. The oxidation of microtubules leads to its cleavage and the subsequent decline in MMP.
Collapse
Affiliation(s)
- Jili Qian
- Unit of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qianping Gao
- Unit of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
38
|
Abstract
Necrosis is a hallmark of several widespread diseases or their direct complications. In the past decade, we learned that necrosis can be a regulated process that is potentially druggable. RIPK3- and MLKL-mediated necroptosis represents by far the best studied pathway of regulated necrosis. During necroptosis, the release of damage-associated molecular patterns (DAMPs) drives a phenomenon referred to as necroinflammation, a common consequence of necrosis. However, most studies of regulated necrosis investigated cell lines in vitro in a cell autonomous manner, which represents a non-physiological situation. Conclusions based on such work might not necessarily be transferrable to disease states in which synchronized, non-cell autonomous effects occur. Here, we summarize the current knowledge of the pathophysiological relevance of necroptosis in vivo, and in light of this understanding, we reassess the morphological classification of necrosis that is generally used by pathologists. Along these lines, we discuss the paucity of data implicating necroptosis in human disease. Finally, the in vivo relevance of non-necroptotic forms of necrosis, such as ferroptosis, is addressed.
Collapse
Affiliation(s)
- Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
39
|
Silver nanoparticles of different sizes induce a mixed type of programmed cell death in human pancreatic ductal adenocarcinoma. Oncotarget 2017; 9:4675-4697. [PMID: 29435134 PMCID: PMC5797005 DOI: 10.18632/oncotarget.22563] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/13/2017] [Indexed: 12/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma, with the high resistance to chemotherapeutic agents, remains the fourth leading cause of cancer-death in the world. Due to the wide range of biological activity and unique properties, silver nanoparticles (AgNPs) are indicated as agents with potential to overcome barriers involved in chemotherapy failure. Therefore, in our study we decided to assess the ability of AgNPs to kill pancreatic cancer cells, and then to identify the molecular mechanism underlying this effect. Moreover, we evaluated the cytotoxicity of AgNPs against non-tumor cell of the same tissue (hTERT-HPNE cells) for comparison. Our results indicated that AgNPs with size of 2.6 and 18 nm decreased viability, proliferation and caused death of pancreatic cancer cells in a size- and concentration-dependent manner. Ultrastructural analysis identified that cellular uptake of AgNPs resulted in apoptosis, autophagy, necroptosis and mitotic catastrophe. These alterations were associated with increased pro-apoptotic protein Bax and decreased level of anti-apoptotic protein Bcl-2. Moreover, AgNPs significantly elevated the level of tumor suppressor p53 protein as well as necroptosis- and autophagy-related proteins: RIP-1, RIP-3, MLKL and LC3-II, respectively. In addition, we found that PANC-1 cells were more vulnerable to AgNPs-induced cytotoxicity compared to pancreatic non-tumor cells. In conclusion, AgNPs by inducing mixed type of programmed cell death in PANC-1 cells, could provide a new therapeutic strategy to overcome chemoresistance in one of the deadliest human cancer.
Collapse
|
40
|
Tang Q, Chang S, Tian Z, Sun J, Hao L, Wang Z, Zhu S. Efficacy of Indocyanine Green-Mediated Sonodynamic Therapy on Rheumatoid Arthritis Fibroblast-like Synoviocytes. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:2690-2698. [PMID: 28779958 DOI: 10.1016/j.ultrasmedbio.2017.06.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 06/14/2017] [Accepted: 06/29/2017] [Indexed: 06/07/2023]
Abstract
Sonodynamic therapy (SDT) has become a new therapeutic method because of its activation of certain sensitizers by ultrasound. Some studies have reported that indocyanine green (ICG) has the characteristics of a sonosensitizer and favorable fluorescence imaging in synovitis of early inflammatory arthritis. In this study, we aimed to investigate the cytotoxic effect of ICG-mediated SDT on MH7A cells in vitro and the potential mechanisms involved. ICG was found to be taken up mainly in cytoplasm, with maximal uptake in 4 h. Cell viability in ICG-mediated SDT (SDT-0.5 and SDT-1.0) groups decreased significantly to 73.09 ± 1.97% and 54.24 ± 4.66%, respectively; cell apoptosis increased significantly to 26.43 ± 0.91% and 45.93 ± 6.17%, respectively. Moreover, marked loss in mitochondrial membrane potential and greatly increased generation of reactive oxygen species were observed in ICG-mediated SDT groups. Interestingly, the loss in cell viability could be effectively rescued with pretreatment with the reactive oxygen species scavenger N-acetylcysteine. These results indicate that ICG-mediated SDT is cytotoxic to fibroblast-like synoviocytes and is a potential modality for targeted therapy of synovitis in rheumatoid arthritis.
Collapse
Affiliation(s)
- Qin Tang
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ultrasound, Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; State Key Laboratory of Ultrasound Engineering in Medicine Co-founded by Chongqing and Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhonghua Tian
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Hao
- Department of Ultrasound, Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Department of Ultrasound, Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shenyin Zhu
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
41
|
Rengeng L, Qianyu Z, Yuehong L, Zhongzhong P, Libo L. Sonodynamic therapy, a treatment developing from photodynamic therapy. Photodiagnosis Photodyn Ther 2017; 19:159-166. [PMID: 28606724 DOI: 10.1016/j.pdpdt.2017.06.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/19/2017] [Accepted: 06/08/2017] [Indexed: 12/31/2022]
Abstract
Sonodynamic therapy (SDT) as a new non-invasive treatment developed from photodynamic (PDT), it can kill tumor cells specifically and selectively. Moreover, recently studies showed SDT has potential to treat solid tumor, leukemia and atherosclerosis, remove proliferative scars and kill pathogenic microorganism. As SDT has an extensive application prospect, SDT has attracted more and more research recently. This thesis aims to be an informative introduction on SDT. With the assistance of related literature from 2012 to 2016, we introduce the progress of SDT research in six aspects: the therapeutic mechanism of SDT, development of the sound sensitizer, exploration of the size and frequency of ultrasonic energy, application of SDT, comparison between SDT and PDT, and current situation and future of SDT.
Collapse
Affiliation(s)
- Liu Rengeng
- Cancer Center, Southern Medical University, Guangzhou 510315, China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China
| | - Zhang Qianyu
- Cancer Center, Southern Medical University, Guangzhou 510315, China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China
| | - Lang Yuehong
- Cancer Center, Southern Medical University, Guangzhou 510315, China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China
| | - Peng Zhongzhong
- Department of Oncology, Ningbo NO.2 Hospital, Ningbo, Zhejiang, China
| | - Li Libo
- Cancer Center, Southern Medical University, Guangzhou 510315, China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To highlight recent studies that describe novel inflammatory and signaling mechanisms that regulate macrophage death in atherosclerosis. RECENT FINDINGS Macrophages contribute to all stages of atherosclerosis. The traditional dogma states that in homeostatic conditions, macrophages undergo apoptosis and are efficiently phagocytosed to be cleared by a process called efferocytosis. In advanced atherosclerosis, however, defective efferocytosis results in secondary necrosis of these uncleared apoptotic cells, which ultimately contributes to the formation of the characteristic necrotic core and the vulnerable plaque. Here, we outline the different types of lesional macrophage death: apoptosis, autophagic and the newly defined necroptosis (i.e. a type of programmed necrosis). Recent discoveries demonstrate that macrophage necroptosis directly contributes to necrotic core formation and plaque instability. Further, promoting the resolution of inflammation using preresolving mediators has been shown to enhance efferocytosis and decrease plaque vulnerability. Finally, the canonical 'don't eat me' signal CD47 has recently been described as playing an important role in atherosclerotic lesion progression by impairing efficient efferocytosis. Although we have made significant strides in improving our understanding of cell death and clearance mechanisms in atherosclerosis, there still remains unanswered questions as to how these pathways can be harnessed using therapeutics to promote lesion regression and disease stability. SUMMARY Improving our understanding of the mechanisms that regulate macrophage death in atherosclerosis, in particular apoptosis, necroptosis and efferocytosis, will provide novel therapeutic opportunities to resolve atherosclerosis and promote plaque stability.
Collapse
Affiliation(s)
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Canada
- Correspondence to: Denuja Karunakaran, PhD or Katey J Rayner, PhD, Cardiometabolic microRNA Laboratory, University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, K1Y 4W7. ; or
| | - Denuja Karunakaran
- University of Ottawa Heart Institute, Ottawa, Canada
- Correspondence to: Denuja Karunakaran, PhD or Katey J Rayner, PhD, Cardiometabolic microRNA Laboratory, University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, K1Y 4W7. ; or
| |
Collapse
|
43
|
Berberine-sonodynamic therapy induces autophagy and lipid unloading in macrophage. Cell Death Dis 2017; 8:e2558. [PMID: 28102849 PMCID: PMC5386349 DOI: 10.1038/cddis.2016.354] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 09/19/2016] [Accepted: 09/27/2016] [Indexed: 01/28/2023]
Abstract
Impaired autophagy in macrophages accompanies the progression of atherosclerosis and contributes to lipid loading in plaques and ineffective lipid degradation. Therefore, evoking autophagy and its associated cholesterol efflux may provide a therapeutic treatment for atherosclerosis. In the present study, berberine-mediated sonodynamic therapy (BBR-SDT) was used to induce autophagy and cholesterol efflux in THP-1 macrophages and derived foam cells. Following BBR-SDT, autophagy was increased in the macrophages, autophagy resistance in the foam cells was prevented, and cholesterol efflux was induced. The first two effects were blocked by the reactive oxygen species scavenger, N-acetyl cysteine. BBR-SDT also reduced the phosphorylation of Akt and mTOR, two key molecules in the PI3K/AKT/mTOR signaling pathway, which is responsible for inducing autophagy. Correspondingly, treatment with the autophagy inhibitor, 3-methyladenine, or the PI3K inhibitor, LY294002, abolished the autophagy-induced effects of BBR-SDT. Furthermore, induction of cholesterol efflux by BBR-SDT was reversed by an inhibition of autophagy by 3-methyladenine or by a small interfering RNA targeting Atg5. Taken together, these results demonstrate that BBR-SDT effectively promotes cholesterol efflux by increasing reactive oxygen species generation, and this subsequently induces autophagy via the PI3K/AKT/mTOR signaling pathway in both ‘normal' macrophages and lipid-loaded macrophages (foam cells). Thus, BBR-SDT may be a promising atheroprotective therapy to inhibit the progression of atherosclerosis and should be further studied.
Collapse
|
44
|
Li X, Zhang X, Zheng L, Kou J, Zhong Z, Jiang Y, Wang W, Dong Z, Liu Z, Han X, Li J, Tian Y, Zhao Y, Yang L. Hypericin-mediated sonodynamic therapy induces autophagy and decreases lipids in THP-1 macrophage by promoting ROS-dependent nuclear translocation of TFEB. Cell Death Dis 2016; 7:e2527. [PMID: 28005078 PMCID: PMC5260986 DOI: 10.1038/cddis.2016.433] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/29/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022]
Abstract
Lipid catabolism disorder is the primary cause of atherosclerosis. Transcription factor EB (TFEB) prevents atherosclerosis by activating macrophage autophagy to promote lipid degradation. Hypericin-mediated sonodynamic therapy (HY-SDT) has been proved non-invasively inducing THP-1-derived macrophage apoptosis; however, it is unknown whether macrophage autophagy could be triggered by HY-SDT to influence cellular lipid catabolism via regulating TFEB. Here, we report that HY-SDT resulted in the time-dependent THP-1-derived macrophage autophagy activation through AMPK/AKT/mTOR pathway. Besides, TFEB nuclear translocation in macrophage was triggered by HY-SDT to promote autophagy activation and lysosome regeneration which enhanced lipid degradation in response to atherogenic lipid stressors. Moreover, following HY-SDT, the ABCA1 expression level was increased to promote lipid efflux in macrophage, and the expression levels of CD36 and SR-A were decreased to inhibit lipid uptake, both of which were prevented by TFEB knockdown. These results indicated that TFEB nuclear translocation activated by HY-SDT was not only the key regulator of autophagy activation and lysosome regeneration in macrophage to promote lipolysis, but also had a crucial role in reverse cholesterol transporters to decrease lipid uptake and increase lipid efflux. Reactive oxygen species (ROS) were adequately generated in macrophage by HY-SDT. Further, ROS scavenger N-acetyl-l-cysteine abolished HY-SDT-induced TFEB nuclear translocation and autophagy activation, implying that ROS were the primary upstream factors responsible for these effects during HY-SDT. In summary, our data indicate that HY-SDT decreases lipid content in macrophage by promoting ROS-dependent nuclear translocation of TFEB to influence consequent autophagy activation and cholesterol transporters. Thus, HY-SDT may be beneficial for atherosclerosis via TFEB regulation to ameliorate lipid overload in atherosclerotic plaques.
Collapse
Affiliation(s)
- Xuesong Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Xin Zhang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Longbin Zheng
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiayuan Kou
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Zhaoyu Zhong
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Yueqing Jiang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Wei Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, China
| | - Zhongni Liu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaobo Han
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Jing Li
- Department of Electron Microscopic Center, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Ye Tian
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China.,Division of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Liming Yang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|