1
|
Li Y, Li Z, Ren Y, Lei Y, Yang S, Shi Y, Peng H, Yang W, Guo T, Yu Y, Xiong Y. Mitochondrial-derived peptides in cardiovascular disease: Novel insights and therapeutic opportunities. J Adv Res 2024; 64:99-115. [PMID: 38008175 PMCID: PMC11464474 DOI: 10.1016/j.jare.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Mitochondria-derived peptides (MDPs) represent a recently discovered family of peptides encoded by short open reading frames (ORFs) found within mitochondrial genes. This group includes notable members including humanin (HN), mitochondrial ORF of the 12S rDNA type-c (MOTS-c), and small humanin-like peptides 1-6 (SHLP1-6). MDPs assume pivotal roles in the regulation of diverse cellular processes, encompassing apoptosis, inflammation, and oxidative stress, which are all essential for sustaining cellular viability and normal physiological functions. Their emerging significance extends beyond this, prompting a deeper exploration into their multifaceted roles and potential applications. AIM OF REVIEW This review aims to comprehensively explore the biogenesis, various types, and diverse functions of MDPs. It seeks to elucidate the central roles and underlying mechanisms by which MDPs participate in the onset and development of cardiovascular diseases (CVDs), bridging the connections between cell apoptosis, inflammation, and oxidative stress. Furthermore, the review highlights recent advancements in clinical research related to the utilization of MDPs in CVD diagnosis and treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW MDPs levels are diminished with aging and in the presence of CVDs, rendering them potential new indicators for the diagnosis of CVDs. Also, MDPs may represent a novel and promising strategy for CVD therapy. In this review, we delve into the biogenesis, various types, and diverse functions of MDPs. We aim to shed light on the pivotal roles and the underlying mechanisms through which MDPs contribute to the onset and advancement of CVDs connecting cell apoptosis, inflammation, and oxidative stress. We also provide insights into the current advancements in clinical research related to the utilization of MDPs in the treatment of CVDs. This review may provide valuable information with MDPs for CVD diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Ying Lei
- School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Silong Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuqi Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Han Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Weijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Tiantian Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
2
|
Elbedwehy AM, Wu J, Na HK, Baek A, Jung H, Kwon IH, Lee SW, Kim JH, Lee TG. ROS-responsive charge reversal mesoporous silica nanoparticles as promising drug delivery system for neovascular retinal diseases. J Control Release 2024; 373:224-239. [PMID: 39002796 DOI: 10.1016/j.jconrel.2024.07.022] [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: 11/25/2023] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Intravitreal injection of biodegradable implant drug carriers shows promise in reducing the injection frequency for neovascular retinal diseases. However, current intravitreal ocular devices have limitations in adjusting drug release rates for individual patients, thereby affecting treatment effectiveness. Accordingly, we developed mesoporous silica nanoparticles (MSNs) featuring a surface that reverse its charge in response to reactive oxygen species (ROS) for efficient delivery of humanin peptide (HN) to retinal epithelial cells (ARPE-19). The MSN core, designed with a pore size of 2.8 nm, ensures a high HN loading capacity 64.4% (w/w). We fine-tuned the external surface of the MSNs by incorporating 20% Acetyl-L-arginine (Ar) to create a partial positive charge, while 80% conjugated thioketal (TK) methoxy polyethylene glycol (mPEG) act as ROS gatekeeper. Ex vivo experiments using bovine eyes revealed the immobilization of Ar-MSNs-TK-PEG (mean zeta potential: 2 mV) in the negatively charged vitreous. However, oxidative stress reversed the surface charge to -25 mV by mPEG loss, facilitating the diffusion of the nanoparticles impeded with HN. In vitro studies showed that ARPE-19 cells effectively internalize HN-loaded Ar-MSNs-TK, subsequently releasing the peptide, which offered protection against oxidative stress-induced apoptosis, as evidenced by reduced TUNEL and caspase3 activation. The inhibition of retinal neovascularization was further validated in an in vivo oxygen-induced retinopathy (OIR) mouse model.
Collapse
Affiliation(s)
- Ahmed M Elbedwehy
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea; Nanotechnology Center, Mansoura University, Mansoura 35516, Egypt
| | - Jun Wu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hee-Kyung Na
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Ahruem Baek
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Haejin Jung
- Flow Cytometry Core Facility of Research Solution Center, Institute of Basic Science, Daejeon 34126, Republic of Korea
| | - Ik Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Sang Won Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Ophthalmology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Tae Geol Lee
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea.
| |
Collapse
|
3
|
Kannoujia J, Chirra N, Rodda R, Shaikh A, Maddala N, Kantevari S, Sripadi P. Identification and characterization of impurities in an insecticide, commercial chlorantraniliprole using liquid chromatography-tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9729. [PMID: 38456250 DOI: 10.1002/rcm.9729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 03/09/2024]
Abstract
RATIONALE Ensuring the global safety and effectiveness of agrochemicals has become imperative. An in-depth understanding of impurity profiles of products is crucial, especially for high-demand agrochemicals, where impurities may be more toxic and persistent than original agrochemicals. This study focuses on the detection and identification of impurities in a commercial chlorantraniliprole (CAP), an anthranilic diamide class broad-spectrum insecticide. METHODS Commercial CAP was collected from an agrochemical supplier in India and was analyzed using a high-performance liquid chromatography-photodiode array (HPLC-PDA) (Agilent 1260; wavelength, 220 nm) with a Zorbax RP SB-C18 (250 × 4.6 mm, 5 μm) column and liquid chromatography-mass spectrometry (LC-MS) (Agilent 6545 quadrupole time of flight (Q-TOF)) techniques to identify the impurities. The impurities were isolated by preparative HPLC using a Zorbax-DB C18 (250 × 9.4 mm, 5 μm) column. liquid chromatography- tandem mass spectrometry (LC-MS/MS) experiments (Q-TOF) were performed on CAP and its impurities to obtain their structural data. RESULTS HPLC-PDA analysis of CAP showed four major impurities (IM-1 to IM-4) ranging from 0.76% to 4.1%. The positive ion electrospray ionization (ESI) mass spectra of CAP and its impurities showed dominant [M + H]+ ions in addition to [M + Na]+ , [M + K]+ , and [2M + Na]+ ions. High-resolution mass spectrometry (HRMS) data provided the elemental composition of the compounds, and isotopic distribution patterns revealed the number of Cl and/or Br atoms present in them. The structures of impurities were proposed based on the LC-MS/MS) data and further confirmed by nuclear magnetic resonance (NMR) data on isolated impurities/synthesis. CONCLUSION The quality and impurities of CAP, a popular insecticide, must be assessed and described for its efficacy and safety. In this study, four impurities of CAP were detected using HPLC and successfully characterized using LC-HRMS, LC-MS/MS, and NMR data. The method is useful for verifying the purity of CAP as well as helping in the identification of its possible impurities.
Collapse
Affiliation(s)
- Jyoti Kannoujia
- Department of Analytical and Structural Chemistry, Centre for Mass Spectrometry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nagaraju Chirra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Fluro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Ramesh Rodda
- Department of Analytical and Structural Chemistry, Centre for Mass Spectrometry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Asif Shaikh
- Department of Analytical and Structural Chemistry, Centre for Mass Spectrometry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nagaraju Maddala
- Department of Analytical and Structural Chemistry, Centre for Mass Spectrometry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Srinivas Kantevari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Fluro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Prabhakar Sripadi
- Department of Analytical and Structural Chemistry, Centre for Mass Spectrometry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [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] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
5
|
Kwek MSY, Thangaveloo M, Madden LE, Phillips ARJ, Becker DL. Targeting Cx43 to Reduce the Severity of Pressure Ulcer Progression. Cells 2023; 12:2856. [PMID: 38132176 PMCID: PMC10741864 DOI: 10.3390/cells12242856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
In the skin, repeated incidents of ischemia followed by reperfusion can result in the breakdown of the skin and the formation of a pressure ulcer. Here we gently applied paired magnets to the backs of mice to cause ischemia for 1.5 h and then removed them to allow reperfusion. The sterile inflammatory response generated within 4 h causes a stage 1 pressure ulcer with an elevation of the gap junction protein Cx43 in the epidermis. If this process is repeated the insult will result in a more severe stage 2 pressure ulcer with a breakdown of the epidermis 2-3 days later. After a single pinch, the elevation of Cx43 in the epidermis is associated with the inflammatory response with an increased number of neutrophils, HMGB1 (marker of necrosis) and RIP3 (responsible for necroptosis). Delivering Cx43 specific antisense oligonucleotides sub-dermally after a single insult, was able to significantly reduce the elevation of epidermal Cx43 protein expression and reduce the number of neutrophils and prevent the elevation of HMGB1 and RIP3. In a double pinch model, the Cx43 antisense treatment was able to reduce the level of inflammation, necroptosis, and the extent of tissue damage and progression to an open wound. This approach may be useful in reducing the progression of stage 1 pressure ulcers to stage 2.
Collapse
Affiliation(s)
- Milton Sheng Yi Kwek
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore (M.T.); (L.E.M.)
- Skin Research Institute Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore
| | - Moogaambikai Thangaveloo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore (M.T.); (L.E.M.)
- Skin Research Institute Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore
| | - Leigh E. Madden
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore (M.T.); (L.E.M.)
- Skin Research Institute Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore
| | | | - David L. Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore (M.T.); (L.E.M.)
- Skin Research Institute Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore 308232, Singapore
| |
Collapse
|
6
|
Zhao Q, Cai MM, Li D, Zhao BY, Zhou SS, Wu ZR, Shi YJ, Su L. S14G-humanin confers cardioprotective effects against chronic adrenergic and pressure overload-induced heart failure in mice. Heliyon 2023; 9:e21892. [PMID: 38045183 PMCID: PMC10692773 DOI: 10.1016/j.heliyon.2023.e21892] [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: 01/03/2023] [Revised: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
S14G-humanin (HNG), an analog of the mitochondria-derived peptide humanin, has demonstrated protective effects against various cardiovascular diseases. However, the specific pharmacological effects of HNG in heart failure (HF) have not been previously reported. Therefore, in this study, we aimed to investigate the potential protective effect of HNG in HF using a mouse model. HF was induced in mice through intraperitoneal injection of isoproterenol or transverse aortic constriction, followed by separate administration of HNG to assess its therapeutic impact. Our results revealed that HNG treatment significantly delayed the onset of cardiac dysfunction and structural remodeling in the HF mouse model. Furthermore, HNG administration was associated with reduced infiltration of inflammatory cells, improved myocardial fibrosis, and attenuation of cardiomyocyte apoptosis in the treated cardiac tissues. Additionally, we identified the involvement of the transforming growth factor-beta signaling pathway in the beneficial effects of HNG in isoproterenol-induced HF mice. Collectively, these findings underscore the therapeutic potential of HNG in preventing the progression of HF, as demonstrated in two distinct HF mouse models.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ming-Ming Cai
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Dan Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bin-Yi Zhao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Shuang-Shan Zhou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhen-Ru Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yu-Jun Shi
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Li Su
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
7
|
Qu Y, Liu Y, Zhang H. ALDH2 activation attenuates oxygen-glucose deprivation/reoxygenation-induced cell apoptosis, pyroptosis, ferroptosis and autophagy. Clin Transl Oncol 2023; 25:3203-3216. [PMID: 37103763 DOI: 10.1007/s12094-023-03190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/04/2023] [Indexed: 04/28/2023]
Abstract
PURPOSE It is previously reported that aldehyde dehydrogenase 2 family member (ALDH2) shows neuroprotective effects in cerebral ischemia/reperfusion injury. However, whether the protective effects are through mediating the programmed cell death is yet to be fully elucidated. METHODS In vitro oxygen-glucose deprivation/reoxygenation (OGD/R) model was established in HT22 cells and mouse cortical neurons. Subsequently, ALDH2 expression were assessed by qRT-PCR and western blot. The methylation status was examined by methylation-specific PCR (MS-PCR). Then, ALDH2 expression was promoted and suppressed to explore the role of ALDH2 in OGD/R-treated cells. CCK-8 assay was applied to detect cell viability, and flow cytometry was applied to evaluate cell apoptosis. Western blot was applied to detect the apoptosis-related proteins (Caspase 3, Bcl-2 and Bax), necroptosis-related proteins (RIP3 and MLKL), pyroptosis-related proteins (NLRP3 and GSDMD), ferroptosis-related protein (ACSL4 and GPX4), and autophagy-related proteins (LC3B, and p62). IL-1β and IL-18 production was evaluated by ELISA assay. Reactive oxygen species production and Fe2+ content were evaluated by the corresponding detection kit. RESULTS In OGD/R-treated cells, ALDH2 expression was decreased, which was due to the hypermethylation of ALDH2 in the promoter region. ALDH2 overexpression improved cell viability and ALDH2 knockdown suppressed cell viability in OGD/R-treated cells. We also found that ALDH2 overexpression attenuated OGD/R-induced cell apoptosis, pyroptosis, ferroptosis and autophagy, while ALDH2 knockdown facilitated the OGD/R-induced cell apoptosis, pyroptosis, ferroptosis and autophagy. CONCLUSIONS Collectively, our results implied that ALDH2 attenuated OGD/R-induced cell apoptosis, pyroptosis, ferroptosis and autophagy to promote cell viability in HT22 cells and mouse cortical neurons.
Collapse
Affiliation(s)
- Yun Qu
- Department of Emergency, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Yuanyuan Liu
- Department of Emergency, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Huilong Zhang
- Department of Neurology, Yuhuangding Hospital Affiliated to Qingdao University, No. 20 Yudong Road, Zhifu District, Yantai, 264000, Shandong, China.
| |
Collapse
|
8
|
Yu C, Wang X, Qin J. Effect of necrostatin-1 on sciatic nerve crush injury in rat models. J Orthop Surg Res 2023; 18:74. [PMID: 36717933 PMCID: PMC9885697 DOI: 10.1186/s13018-023-03565-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Necrostatin-1 (Nec-1) is an inhibitor of the receptor interacting protein (RIP)1 kinase, which acts as an inhibitor of necroptosis, a special form of necrosis. In the present study, the effect of Nec-1 on peripheral nerve injury (PNI) was investigated. METHODS The PNI model was established by inducing sciatic nerve injury. Hematoxylin-eosin and immunofluorescence staining techniques were used to assess the extent of injury to nerve fibers and necrosis of Schwann cells (SCs). Western blotting was performed to detect the expression of necroptosis-related factors (RIP1 and RIP3). The concentrations of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and the oxidative stress-related enzyme malondialdehyde (MDA) were determined to indicate the degree of inflammation and oxidative stress. RESULTS Nec-1 could decrease the degree of peripheral nerve lesions after PNI and protect SCs and axons by inhibiting necroptosis. Furthermore, Nec-1 could reduce necroptosis by inhibiting RIP1 and effectively reduce inflammation and reactive oxygen species production at the early stage of PNI. CONCLUSIONS Alleviation of necroptosis by Nec-1 may provide new insights into therapies for the early stages of peripheral nerve repair after PNI.
Collapse
Affiliation(s)
- Chen Yu
- grid.89957.3a0000 0000 9255 8984Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Avenue, Nanjing, 210000 Jiangsu China
| | - Xiaoxu Wang
- grid.412017.10000 0001 0266 8918Department of Orthopaedics, The Second Hospital, University of South China, Hengyang, 421000 Hunan China
| | - Jian Qin
- grid.89957.3a0000 0000 9255 8984Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Avenue, Nanjing, 210000 Jiangsu China
| |
Collapse
|
9
|
FU JIAWEI, WU CHUNSHUAI, XU GUANHUA, ZHANG JINLONG, LI YIQIU, JI CHUNYAN, CUI ZHIMING. Role of necroptosis in spinal cord injury and its therapeutic implications. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
10
|
He Y, Ying J, Tang J, Zhou R, Qu H, Qu Y, Mu D. Neonatal Arterial Ischaemic Stroke: Advances in Pathologic Neural Death, Diagnosis, Treatment, and Prognosis. Curr Neuropharmacol 2022; 20:2248-2266. [PMID: 35193484 PMCID: PMC9890291 DOI: 10.2174/1570159x20666220222144744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal arterial ischaemic stroke (NAIS) is caused by focal arterial occlusion and often leads to severe neurological sequelae. Neural deaths after NAIS mainly include necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. These neural deaths are mainly caused by upstream stimulations, including excitotoxicity, oxidative stress, inflammation, and death receptor pathways. The current clinical approaches to managing NAIS mainly focus on supportive treatments, including seizure control and anticoagulation. In recent years, research on the pathology, early diagnosis, and potential therapeutic targets of NAIS has progressed. In this review, we summarise the latest progress of research on the pathology, diagnosis, treatment, and prognosis of NAIS and highlight newly potential diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
11
|
Chen B, Hang J, Zhao Y, Geng Y, Li X, Gu Z, Li J, Jiang C, Tao L, Yu H. Correlation between Plasma Levels of RIP3 and Acute Ischemic Stroke with Large-Artery Atherosclerosis. Curr Neurovasc Res 2022; 19:30-37. [PMID: 35156583 DOI: 10.2174/1567202619666220214105208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Receptor-interacting serine-threonine protein kinase 3 (RIP3) was previously discovered to be an important medium in the occurrence and development of major atherosclerotic cerebral infarction. However, the role of RIP3 in acute ischemic stroke remains unclear. OBJECTIVE This study aimed to explore the correlation between plasma levels of RIP3 and acute ischemic stroke with large-artery atherosclerosis (LAA). METHODS This prospective study enrolled 116 patients with LAA, 40 healthy controls and 30 acute ischemic stroke patients with small-artery occlusion. The patients with LAA were divided according to the quartile of plasma levels of RIP3. Logistic regression model was used for comparison. The ROC curve was performed to evaluate the predictive value. RESULTS In patients with LAA, the RIP3 levels in patients with poor outcomes as well as neurological deterioration were significantly higher than those with good outcomes (P < 0.001) and without neurological deterioration (P = 0.014) respectively. Patients in the highest levels of plasma RIP3 quartile were more likely to have neurological deterioration (OR, 11.07; 95% CI, 1.990-61.582) and poor outcomes (OR, 35.970; 95% CI, 5.392-239.980) compared with the lowest. The optimal cut-off value for neurological deterioration was 1127.75 pg/mL (specificity, 66.7%; sensitivity, 69.2%), that for poor prognosis was 1181.82 pg/mL (specificity, 89.7%; sensitivity, 62.1%). CONCLUSION Elevated levels of plasma RIP3 were significantly associated with neurological deterioration and poor prognosis in patients with LAA. Significant increase in plasma RIP3 levels can predict neurological deterioration and poor prognosis of these patients.
Collapse
Affiliation(s)
- Beilei Chen
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jing Hang
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Yuanyuan Zhao
- Department of Neurology, Bazhong Central Hospital, Bazhong, Sichuang
| | - Yang Geng
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Xiaobo Li
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Zhie Gu
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jun Li
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Chao Jiang
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Luhang Tao
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Hailong Yu
- Clinical Medical College of Yangzhou University, Yangzhou, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
12
|
Emerging immune and cell death mechanisms in stroke: Saponins as therapeutic candidates. Brain Behav Immun Health 2021; 9:100152. [PMID: 34589895 PMCID: PMC8474497 DOI: 10.1016/j.bbih.2020.100152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The complexity of the ischemic cascade is based on the integrated crosstalk of every cell type in the neurovascular unit. Depending on the features of the ischemic insult, several cell death mechanisms are triggered, such as apoptosis, necroptosis, ferroptosis/oxytosis, ETosis or pyroptosis, leading to reactive astrogliosis. However, emerging evidence demonstrates a dual role for the immune system in stroke pathophysiology, where it exerts both detrimental and also beneficial functions. In this review, we discuss the relevance of several cell death modalities and the dual role of the immune system in stroke pathophysiology. We also provide an overview of some emerging immunomodulatory therapeutic strategies, amongst which saponins, which are promising candidates that exert multiple pharmacological effects. Several cell death mechanisms coexist in stroke pathophysiology. Neurons are more vulnerable to necroptosis than glial cells. Inhibitors of receptor-interacting protein kinases and of ferroptosis induce neuroprotection. Saponins exert modulatory effects on inflammation and neuronal cell death in stroke.
Collapse
|
13
|
Jantas D, Lasoń W. Preclinical Evidence for the Interplay between Oxidative Stress and RIP1-Dependent Cell Death in Neurodegeneration: State of the Art and Possible Therapeutic Implications. Antioxidants (Basel) 2021; 10:antiox10101518. [PMID: 34679652 PMCID: PMC8532910 DOI: 10.3390/antiox10101518] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are the most frequent chronic, age-associated neurological pathologies having a major impact on the patient’s quality of life. Despite a heavy medical, social and economic burden they pose, no causative treatment is available for these diseases. Among the important pathogenic factors contributing to neuronal loss during neurodegeneration is elevated oxidative stress resulting from a disturbed balance between endogenous prooxidant and antioxidant systems. For many years, it was thought that increased oxidative stress was a cause of neuronal cell death executed via an apoptotic mechanism. However, in recent years it has been postulated that rather programmed necrosis (necroptosis) is the key form of neuronal death in the course of neurodegenerative diseases. Such assumption was supported by biochemical and morphological features of the dying cells as well as by the fact that various necroptosis inhibitors were neuroprotective in cellular and animal models of neurodegenerative diseases. In this review, we discuss the relationship between oxidative stress and RIP1-dependent necroptosis and apoptosis in the context of the pathomechanism of neurodegenerative disorders. Based on the published data mainly from cellular models of neurodegeneration linking oxidative stress and necroptosis, we postulate that administration of multipotential neuroprotectants with antioxidant and antinecroptotic properties may constitute an efficient pharmacotherapeutic strategy for the treatment of neurodegenerative diseases.
Collapse
|
14
|
Ouyang Y, Wen L, Armstrong JA, Chvanov M, Latawiec D, Cai W, Awais M, Mukherjee R, Huang W, Gough PJ, Bertin J, Tepikin AV, Sutton R, Criddle DN. Protective Effects of Necrostatin-1 in Acute Pancreatitis: Partial Involvement of Receptor Interacting Protein Kinase 1. Cells 2021; 10:1035. [PMID: 33925729 PMCID: PMC8145347 DOI: 10.3390/cells10051035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
Acute pancreatitis (AP) is a severe and potentially fatal disease caused predominantly by alcohol excess and gallstones, which lacks a specific therapy. The role of Receptor-Interacting Protein Kinase 1 (RIPK1), a key component of programmed necrosis (Necroptosis), is unclear in AP. We assessed the effects of RIPK1 inhibitor Necrostatin-1 (Nec-1) and RIPK1 modification (RIPK1K45A: kinase dead) in bile acid (TLCS-AP), alcoholic (FAEE-AP) and caerulein hyperstimulation (CER-AP) mouse models. Involvement of collateral Nec-1 target indoleamine 2,3-dioxygenase (IDO) was probed with the inhibitor Epacadostat (EPA). Effects of Nec-1 and RIPK1K45A were also compared on pancreatic acinar cell (PAC) fate in vitro and underlying mechanisms explored. Nec-1 markedly ameliorated histological and biochemical changes in all models. However, these were only partially reduced or unchanged in RIPK1K45A mice. Inhibition of IDO with EPA was protective in TLCS-AP. Both Nec-1 and RIPK1K45A modification inhibited TLCS- and FAEE-induced PAC necrosis in vitro. Nec-1 did not affect TLCS-induced Ca2+ entry in PACs, however, it inhibited an associated ROS elevation. The results demonstrate protective actions of Nec-1 in multiple models. However, RIPK1-dependent necroptosis only partially contributed to beneficial effects, and actions on targets such as IDO are likely to be important.
Collapse
Affiliation(s)
- Yulin Ouyang
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (Y.O.); (M.C.); (A.V.T.)
- Brain Cognition and Brain Disease Institute, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Li Wen
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Jane A. Armstrong
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Michael Chvanov
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (Y.O.); (M.C.); (A.V.T.)
| | - Diane Latawiec
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Wenhao Cai
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Mohammad Awais
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Rajarshi Mukherjee
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Wei Huang
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - Peter J. Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA; (P.J.G.); (J.B.)
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA; (P.J.G.); (J.B.)
| | - Alexei V. Tepikin
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (Y.O.); (M.C.); (A.V.T.)
| | - Robert Sutton
- Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (L.W.); (J.A.A.); (D.L.); (W.C.); (M.A.); (R.M.); (W.H.); (R.S.)
| | - David N. Criddle
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK; (Y.O.); (M.C.); (A.V.T.)
| |
Collapse
|
15
|
Stark K, Goncharov T, Varfolomeev E, Xie L, Ngu H, Peng I, Anderson KR, Verschueren E, Choi M, Kirkpatrick DS, Easton A, Webster JD, McKenzie BS, Vucic D, Bingol B. Genetic inactivation of RIP1 kinase activity in rats protects against ischemic brain injury. Cell Death Dis 2021; 12:379. [PMID: 33828080 PMCID: PMC8026634 DOI: 10.1038/s41419-021-03651-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 02/01/2023]
Abstract
RIP1 kinase-mediated inflammatory and cell death pathways have been implicated in the pathology of acute and chronic disorders of the nervous system. Here, we describe a novel animal model of RIP1 kinase deficiency, generated by knock-in of the kinase-inactivating RIP1(D138N) mutation in rats. Homozygous RIP1 kinase-dead (KD) rats had normal development, reproduction and did not show any gross phenotypes at baseline. However, cells derived from RIP1 KD rats displayed resistance to necroptotic cell death. In addition, RIP1 KD rats were resistant to TNF-induced systemic shock. We studied the utility of RIP1 KD rats for neurological disorders by testing the efficacy of the genetic inactivation in the transient middle cerebral artery occlusion/reperfusion model of brain injury. RIP1 KD rats were protected in this model in a battery of behavioral, imaging, and histopathological endpoints. In addition, RIP1 KD rats had reduced inflammation and accumulation of neuronal injury biomarkers. Unbiased proteomics in the plasma identified additional changes that were ameliorated by RIP1 genetic inactivation. Together these data highlight the utility of the RIP1 KD rats for target validation and biomarker studies for neurological disorders.
Collapse
Affiliation(s)
- Kimberly Stark
- grid.418158.10000 0004 0534 4718Department of Neuroscience, Genentech, South San Francisco, 94080 CA USA
| | - Tatiana Goncharov
- grid.418158.10000 0004 0534 4718Department of Early Discovery Biochemistry, Genentech, South San Francisco, 94080 CA USA
| | - Eugene Varfolomeev
- grid.418158.10000 0004 0534 4718Department of Early Discovery Biochemistry, Genentech, South San Francisco, 94080 CA USA
| | - Luke Xie
- grid.418158.10000 0004 0534 4718Department of Biomedical Imaging, Genentech, South San Francisco, 94080 CA USA
| | - Hai Ngu
- grid.418158.10000 0004 0534 4718Department of Pathology, Genentech, South San Francisco, 94080 CA USA
| | - Ivan Peng
- grid.418158.10000 0004 0534 4718Department of Translational Immunology, Genentech, South San Francisco, 94080 CA USA
| | - Keith R. Anderson
- grid.418158.10000 0004 0534 4718Department of Molecular Biology, Genentech, South San Francisco, 94080 CA USA
| | - Erik Verschueren
- grid.418158.10000 0004 0534 4718Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, 94080 CA USA
| | - Meena Choi
- grid.418158.10000 0004 0534 4718Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, 94080 CA USA
| | - Donald S. Kirkpatrick
- grid.418158.10000 0004 0534 4718Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, 94080 CA USA
| | - Amy Easton
- grid.418158.10000 0004 0534 4718Department of Neuroscience, Genentech, South San Francisco, 94080 CA USA
| | - Joshua D. Webster
- grid.418158.10000 0004 0534 4718Department of Pathology, Genentech, South San Francisco, 94080 CA USA
| | - Brent S. McKenzie
- grid.418158.10000 0004 0534 4718Department of Translational Immunology, Genentech, South San Francisco, 94080 CA USA
| | - Domagoj Vucic
- grid.418158.10000 0004 0534 4718Department of Early Discovery Biochemistry, Genentech, South San Francisco, 94080 CA USA
| | - Baris Bingol
- grid.418158.10000 0004 0534 4718Department of Neuroscience, Genentech, South San Francisco, 94080 CA USA
| |
Collapse
|
16
|
Xin N, Lu J, Zhou Y, Cheng Y. Dexamethasone Protects Against Ischaemic Brain Injury via Inhibiting the pAkt Signalling Pathway Through Increasing Hap1. Neurotox Res 2021; 39:191-197. [PMID: 32876919 DOI: 10.1007/s12640-020-00255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 10/23/2022]
Abstract
Dexamethasone has been reported to reduce the infarct volume and protect neurological function after ischaemic injury, but the mechanism of Dex in brain injury is not clear. We aimed to study the mechanism by which dexamethasone protects against ischaemic brain injury. Western blotting was to detected the expression of Hap1,TrkB, Akt and Erk; TTC staining to analyse ischemic volume; neurological deficit evaluation to test degree of ischemic injury; immunofluorescence staining to analyse the distribution of Hap1; and the MCAO model was used to study these processes. All data are expressed as the means ± SEM and were analysed by GraphPad Prism 6. P < 0.05 was considered statistically significant. After dexamethasone (Dex) treatment, Hap1 levels were increased and peaked at 2 days; then, we found that body weight was decreased in Hap1-/+ mice. Further study showed that Dex treatment reduced the ischaemic volume and improved neurological function. Finally, we showed that Hap1 regulated the levels of pTrkB, pAkt and pErk 1/2 in ischaemic injury after Dex treatment. Our data suggest that dexamethasone protects against ischaemic brain injury by inhibiting the pAkt signalling pathway through increasing Hap1.
Collapse
Affiliation(s)
- Ning Xin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, 221000, Jiangsu Province, China
| | - Jun Lu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, 221000, Jiangsu Province, China
| | - Yanlong Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, 221000, Jiangsu Province, China
| | - Yanbo Cheng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, 221000, Jiangsu Province, China.
| |
Collapse
|
17
|
Jia Y, Swerdloff RS, Lue Y, Dai-Ju J, Surampudi P, Cohen P, Wang C. The IL-27 component EBI-3 and its receptor subunit IL-27Rα are essential for the cytoprotective action of humanin on male germ cells†. Biol Reprod 2021; 104:717-730. [PMID: 33330922 PMCID: PMC8527998 DOI: 10.1093/biolre/ioaa225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/19/2020] [Accepted: 12/08/2020] [Indexed: 12/28/2022] Open
Abstract
Humanin (HN) is a mitochondrial-derived peptide that protects many cells/tissues from damage. We previously demonstrated that HN reduces stress-induced male germ cell apoptosis in rodents. HN action in neuronal cells is mediated through its binding to a trimeric cell membrane receptor composed of glycoprotein 130 (gp130), IL-27 receptor subunit (IL-27R, also known as WSX-1/TCCR), and ciliary neurotrophic factor receptor subunit (CNTFR). The mechanisms of HN action in testis remain unclear. We demonstrated in ex-vivo seminiferous tubules culture that HN prevented heat-induced germ cell apoptosis was blocked by specific anti-IL-27R, anti-gp130, and anti-EBI-3, but not by anti-CNTFR antibodies significantly. The cytoprotective action of HN was studied by using groups of il-27r-/- or ebi-3-/- mice administered the following treatment: (1) vehicle; (2) a single intraperitoneal (IP) injection of HN peptide; (3) testicular hyperthermia; and (4) testicular hyperthermia plus HN. We demonstrated that HN inhibited heat-induced germ cell apoptosis in wildtype but not in il-27r-/- or ebi-3-/- mice. HN restored heat-suppressed STAT3 phosphorylation in wildtype but not il-27r-/- or ebi-3-/- mice. Dot blot analyses showed the direct interaction of HN with IL-27R or EBI-3 peptide. Immunofluorescence staining showed the co-localization of IL-27R with HN and gp130 in Leydig cells and germ cells. We conclude that the anti-apoptotic effects of HN in mouse testes are mediated through interaction with EBI-3, IL-27R, and activation of gp130, whereas the role of CNTFR needs further studies. This suggests a multicomponent tissue-specific receptor for HN in the testis and links HN action with the IL-12/IL-27 family of cytokines.
Collapse
Affiliation(s)
- Yue Jia
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ronald S Swerdloff
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - YanHe Lue
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jenny Dai-Ju
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Prasanth Surampudi
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pinchas Cohen
- USC Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, USA
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, The Lundquist Research Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
18
|
Zanetti LC, Weinlich R. Necroptosis, the Other Main Caspase-Independent Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:123-138. [PMID: 34370290 DOI: 10.1007/978-3-030-62026-4_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The past decades witnessed the discovery of novel modes of cell death, such as ferroptosis, pyroptosis and necroptosis, all of them presenting common necrotic traits. In this chapter, we revisit the early discoveries that unveiled necroptosis as a distinct cell death mechanism. We describe necroptosis, its main regulators and their role in maintaining cellular homeostasis and in the disease state. We conclude by discussing its phenotypic similarities with ferroptosis and the possible crosstalk between these pathways.
Collapse
Affiliation(s)
- Larissa C Zanetti
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil.
| | - Ricardo Weinlich
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Dose-dependent effects of necrostatin-1 supplementation to tissue culture media of young porcine islets. PLoS One 2020; 15:e0243506. [PMID: 33284818 PMCID: PMC7721208 DOI: 10.1371/journal.pone.0243506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/20/2020] [Indexed: 12/23/2022] Open
Abstract
Previous studies have shown that necrostatin-1 (Nec-1) supplementation improved the viability of murine islets following exposure to nitric oxide, increased the survival of human islets during hypoxic culture, and augmented the maturation of pre-weaned porcine islets (PPIs) after 7 days of tissue culture. A limitation of these studies is that only one concentration of Nec-1 was used, and no studies have determined the optimal dose of Nec-1 for PPIs. Thus, the present study examined the effects of Nec-1 on PPIs at four different doses—0, 25, 50, 100, and 200 μM—after 7 days of tissue culture when supplemented on day 3. PPIs were isolated from pancreata of pre-weaned Yorkshire piglets (8–15 days old) and cultured in a specific islet maturation media added with Nec-1 on day 3 of tissue culture at 4 different doses—0, 25, 50, 100, and 200 μM (n = 6 for each dose). After 7 days of tissue culture, islets were assessed for recovery, viability, endocrine cellular content, GLUT2 expression in beta cells, and insulin secretion after glucose challenge. Nec-1 did not affect the viability of both intact islets and dissociated islets cells during tissue culture regardless of doses. Islets cultured in media supplemented with Nec-1 at 100 μM, but not 25, 50, or 200 μM, had a significantly higher recovery, composition of endocrine cells, GLUT2 expression in beta cells, and insulin secretion capacity than control islets cultured in media without Nec-1 supplementation. Moreover, culturing islets in 200 μM Nec-1 supplemented media not only failed to improve the insulin release but resulted in a lower glucose-induced insulin stimulation index compared to islets cultured in media added with 100 μM Nec-1. Xenotransplantation using porcine islets continues to demonstrate scientific advances to justify this area of research. Our findings indicate that Nec-1 supplementation at 100 μM was most effective to enhance the in vitro maturation of PPIs during tissue culture.
Collapse
|
20
|
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: 6.3] [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
|
21
|
Liao S, Apaijai N, Chattipakorn N, Chattipakorn SC. The possible roles of necroptosis during cerebral ischemia and ischemia / reperfusion injury. Arch Biochem Biophys 2020; 695:108629. [PMID: 33068524 DOI: 10.1016/j.abb.2020.108629] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/03/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Cell death is a process consequential to cerebral ischemia and cerebral ischemia/reperfusion (I/R) injury. Recent evidence suggest that necroptosis has been involved in the pathogenesis of ischemic brain injury. The mechanism of necroptosis is initiated by an activation of inflammatory receptors including tumor necrosis factor, toll like receptor, and fas ligands. The signals activate the receptor-interacting protein kinase (RIPK) 1, 3, and a mixed-lineage kinase domain-like pseudokinase (MLKL) to instigate necroptosis. RIPK1 inhibitor, necrostatin-1, was developed, and dramatically reduced brain injury following cerebral ischemia in mice. Consequently, necroptosis could be a novel therapeutic target for stroke, which aims to reduce long-term adverse outcomes after cerebral ischemia. Several studies have been conducted to test the roles of necroptosis on cerebral ischemia and cerebral I/R injury, and the efficacy of necrostatin-1 has been tested in those models. Evidence regarding the roles of necroptosis and the effects of necrostatin-1, from in vitro and in vivo studies, has been summarized and discussed. In addition, other therapeutic managements, involving in necroptosis, are also included in this review. We believe that the insights from this review might clarify the clinical perspective and challenges involved in future stroke treatment by targeting the necroptosis pathway.
Collapse
Affiliation(s)
- Suchan Liao
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
22
|
Jung SE, Ahn JS, Kim YH, Oh HJ, Kim BJ, Ryu BY. Necrostatin-1 improves the cryopreservation efficiency of murine spermatogonial stem cells via suppression of necroptosis and apoptosis. Theriogenology 2020; 158:445-453. [PMID: 33049569 DOI: 10.1016/j.theriogenology.2020.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 08/19/2020] [Accepted: 10/03/2020] [Indexed: 12/21/2022]
Abstract
Cryopreservation of spermatogonial stem cells (SSCs) is important to preserve the lineages of valuable livestock and produce transgenic animals. Although interest in molecular-based cryopreservation methods have been increasing to improve their efficiency, the issue of necroptosis has not yet been considered. Therefore, the purpose of this study was to understand the role of necroptosis using necrostatin-1 (Nec-1), necroptosis inhibitor, in SSC cryopreservation, and to investigate the potential application of Nec-1 as a cryoprotectant. To determine the cryopreservation efficiency of Nec-1, we assessed recovery rate, proliferation potential, cellular membrane damage, RIP1 protein expression, apoptosis, and its mechanism. Stable characterization and functional activity of SSCs was determined via immunofluorescence, RT-qPCR, and in vivo transplantation of SSCs. Our results showed a higher proliferation potential in 50 μM Nec-1 (146.5 ± 16.8%) than in DMSO controls (100.0 ± 3.4%). Furthermore, the cryoprotective effects of Nec-1 were verified by a decrease in RIP1 expression (3.1 ± 0.2-fold vs. 1.3 ± 0.3-fold) and in early apoptosis (4.3 ± 0.8% vs. 2.6 ± 0.1%) compared to DMSO controls. Normal functional activity was observed in the SSCs after cryopreservation with 50 μM Nec-1. In conclusion, necroptosis could be a cause of cryoinjury, and their inhibitor may serve as potential effective cryoprotectant. This study will contribute to establish a molecular-based cryopreservation method, and thereby expanding the use of SSCs into the domestic livestock industry as well as for clinical applications.
Collapse
Affiliation(s)
- Sang-Eun Jung
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do, Republic of Korea
| | - Jin Seop Ahn
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do, Republic of Korea
| | - Yong-Hee Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do, Republic of Korea
| | - Hui-Jo Oh
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do, Republic of Korea.
| |
Collapse
|
23
|
Zhu J, Yang LK, Wang QH, Lin W, Feng Y, Xu YP, Chen WL, Xiong K, Wang YH. NDRG2 attenuates ischemia-induced astrocyte necroptosis via the repression of RIPK1. Mol Med Rep 2020; 22:3103-3110. [PMID: 32945444 PMCID: PMC7453600 DOI: 10.3892/mmr.2020.11421] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 11/20/2019] [Indexed: 12/25/2022] Open
Abstract
Cerebral ischemia results in severe brain damage, and is a leading cause of death and long-term disability. Previous studies have investigated methods to activate astrocytes in order to promote repair in injured brain tissue and inhibit cell death. It has previously been shown that N-myc downstream-regulated gene 2 (NDRG2) was highly expressed in astrocytes and associated with cell activity, but the underlying mechanism is largely unknown. The present study generated NDRG2 conditional knockout (Ndrg2-/-) mice to investigate whether NDRG2 can block ischemia-induced astrocyte necroptosis by suppressing receptor interacting protein kinase 1 (RIPK1) expression. This study investigated astrocyte activity in cerebral ischemia, and identified that ischemic brain injuries could trigger RIP-dependent astrocyte necroptosis. The depletion of NDRG2 was found to accelerate permanent middle cerebral artery occlusion-induced necroptosis in the brain tissue of Ndrg2-/- mice, indicating that NDRG2 may act as a neuroprotector during cerebral ischemic injury. The present study suggested that NDRG2 attenuated astrocytic cell death via the suppression of RIPK1. The pharmacological inhibition of astrocyte necroptosis by necrostatin-1 provided neuroprotection against ischemic brain injuries after NDRG2 knockdown. Therefore, NDRG2 could be considered as a potential target for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Li-Kun Yang
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Qiu-Hong Wang
- Department of Ophthalmology, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Wei Lin
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Yi Feng
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Ye-Ping Xu
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Wei-Liang Chen
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu-Hai Wang
- Department of Neurosurgery, The 101 Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
| |
Collapse
|
24
|
Gunata M, Parlakpinar H. A review of myocardial ischaemia/reperfusion injury: Pathophysiology, experimental models, biomarkers, genetics and pharmacological treatment. Cell Biochem Funct 2020; 39:190-217. [PMID: 32892450 DOI: 10.1002/cbf.3587] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are known to be the most fatal diseases worldwide. Ischaemia/reperfusion (I/R) injury is at the centre of the pathology of the most common cardiovascular diseases. According to the World Health Organization estimates, ischaemic heart disease is the leading global cause of death, causing more than 9 million deaths in 2016. After cardiovascular events, thrombolysis, percutaneous transluminal coronary angioplasty or coronary bypass surgery are applied as treatment. However, after restoring coronary blood flow, myocardial I/R injury may occur. It is known that this damage occurs due to many pathophysiological mechanisms, especially increasing reactive oxygen types. Besides causing cardiomyocyte death through multiple mechanisms, it may be an important reason for affecting other cell types such as platelets, fibroblasts, endothelial and smooth muscle cells and immune cells. Also, polymorphonuclear leukocytes are associated with myocardial I/R damage during reperfusion. This damage may be insufficient in patients with co-morbidity, as it is demonstrated that it can be prevented by various endogenous antioxidant systems. In this context, the resulting data suggest that optimal cardioprotection may require a combination of additional or synergistic multi-target treatments. In this review, we discussed the pathophysiology, experimental models, biomarkers, treatment and its relationship with genetics in myocardial I/R injury. SIGNIFICANCE OF THE STUDY: This review summarized current information on myocardial ischaemia/reperfusion injury (pathophysiology, experimental models, biomarkers, genetics and pharmacological therapy) for researchers and reveals guiding data for researchers, especially in the field of cardiovascular system and pharmacology.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
25
|
Dojo Soeandy C, Elia AJ, Cao Y, Rodgers C, Huang S, Elia AC, Henderson JT. Necroptotic-Apoptotic Regulation in an Endothelin-1 Model of Cerebral Ischemia. Cell Mol Neurobiol 2020; 41:1727-1742. [PMID: 32844322 DOI: 10.1007/s10571-020-00942-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022]
Abstract
The primary forms of cell death seen in ischemic stroke are of two major types: a necrotic/necroptotic form, and an apoptotic form that is frequently seen in penumbral regions of injury. Typically apoptotic versus necroptotic programmed cell death is described as competitive in nature, where necroptosis is often described as playing a backup role to apoptosis. In the present study, we examined the relationship between these two forms of cell death in a murine endothelin-1 model of ischemia-reperfusion injury in wildtype and caspase-3 null mice with and without addition of the pharmacologic RIPK1 phosphorylation inhibitor necrostatin-1. Analyses of ischemic brain injury were performed via both cellular and volumetric assessments, electron microscopy, TUNEL staining, activated caspase-3 and caspase-7 staining, as well as CD11b and F4/80 staining. Inhibition of caspase-3 or RIPK1 phosphorylation demonstrates significant neural protective effects which are non-additive and exhibit significant overlap in protected regions. Interestingly, morphologic analysis of the cortex demonstrates reduced apoptosis following RIPK1 inhibition. Consistent with this, RIPK1 inhibition reduces the levels of both caspase-3 and caspase-7 activation. Additionally, this protection appears independent of secondary inflammatory mediators. Together, these observations demonstrate that the necroptotic protein RIPK1 modifies caspase-3/-7 activity, ultimately resulting in decreased neuronal apoptosis. These findings thus modify the traditional exclusionary view of apoptotic/necroptotic signaling, revealing a new form of interaction between these dominant forms of cell death.
Collapse
Affiliation(s)
- Chesarahmia Dojo Soeandy
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada
| | - Andrew J Elia
- Princess Margaret Cancer Center, University Health Network, 610 University Avenue Rm 7-323, Toronto, ON, M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, 101 College Street Rm 15-701, Toronto, ON, M5G 1L7, Canada
| | - Yanshan Cao
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 1010, Toronto, ON, M5S 3M2, Canada
| | - Christopher Rodgers
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada
| | - Shudi Huang
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada
| | - Andrea C Elia
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada
| | - Jeffrey T Henderson
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
26
|
Catalytically inactive RIP1 and RIP3 deficiency protect against acute ischemic stroke by inhibiting necroptosis and neuroinflammation. Cell Death Dis 2020; 11:565. [PMID: 32703968 PMCID: PMC7378260 DOI: 10.1038/s41419-020-02770-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022]
Abstract
Necroptosis, which is mediated by RIP1/RIP3/MLKL (receptor-interacting protein kinase 1/receptor-interacting protein kinase 3/mixed lineage kinase domain-like protein) signaling, is a critical process in the development of acute ischemic stroke. However, it is unclear precisely how necroptosis promotes the pathogenesis of acute ischemic stroke. In this experimental study in mice, we investigated how necroptosis loss-of-function mice, RIP1 kinase-dead mice, RIP3-deficiency mice, and MLKL-deficiency mice could be protected against cerebral injury after acute ischemic stroke. Insoluble RIP1, RIP3, and MLKL were all detected in the infarct area of the study mice, indicating activation of necroptosis. Two types of RIP1 kinase-dead mutant mice (Rip1K45A/K45A or Rip1Δ/Δ) were used to show that catalytically-inactive RIP1 can decrease the infarct volume and improve neurological function after MCAO/R (middle cerebral artery occlusion/reperfusion). Both Rip3−/− mice and Mlkl−/− mice were protected against acute ischemic stroke. In addition, necroptosis loss-of-function mice showed less inflammatory responses in the infarct area. Therefore, necroptosis and its accompanying inflammatory response can lead to acute injury following ischemia stroke. Our study provides new insight into the pathogenetic mechanisms of acute ischemic stroke, and suggests potential therapeutic targets for neuroprotection.
Collapse
|
27
|
Mifflin L, Ofengeim D, Yuan J. Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat Rev Drug Discov 2020; 19:553-571. [PMID: 32669658 PMCID: PMC7362612 DOI: 10.1038/s41573-020-0071-y] [Citation(s) in RCA: 252] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a key mediator of cell death and inflammation. The unique hydrophobic pocket in the allosteric regulatory domain of RIPK1 has enabled the development of highly selective small-molecule inhibitors of its kinase activity, which have demonstrated safety in preclinical models and clinical trials. Potential applications of these RIPK1 inhibitors for the treatment of monogenic and polygenic autoimmune, inflammatory, neurodegenerative, ischaemic and acute conditions, such as sepsis, are emerging. This article reviews RIPK1 biology and disease-associated mutations in RIPK1 signalling pathways, highlighting clinical trials of RIPK1 inhibitors and potential strategies to mitigate development challenges. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) — a key mediator of cell death and inflammation — is activated in human diseases. Here, Yuan and colleagues discuss current understanding of RIPK1 biology and its association with diseases including inflammatory and autoimmune disorders, neurodegenerative diseases and sepsis. The clinical development of small-molecule RIPK1 inhibitors and associated challenges are discussed.
Collapse
Affiliation(s)
- Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Dimitry Ofengeim
- Rare and Neurologic Disease Research, Sanofi, Framingham, MA, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Liu J, Zhu YM, Guo Y, Lin L, Wang ZX, Gu F, Dong XY, Zhou M, Wang YF, Zhang HL. Inhibition of GSK3β and RIP1K Attenuates Glial Scar Formation Induced by Ischemic Stroke via Reduction of Inflammatory Cytokine Production. Front Pharmacol 2020; 11:812. [PMID: 32595496 PMCID: PMC7303311 DOI: 10.3389/fphar.2020.00812] [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: 07/04/2019] [Accepted: 05/18/2020] [Indexed: 01/05/2023] Open
Abstract
In the chronic phase following ischemic stroke, glial scars can prevent axonal regeneration and the intensification of inflammation. The protective effect of inhibition of glycogen synthase kinase-3β (GSK3β) or receptor-interacting protein 1 kinase (RIP1K) on ischemic stroke has been previously reported. The current study examined the effects of RIP1K and GSK3β on ischemic stroke-induced glial scar formation. To investigate this, we used an in vivo model of ischemic stroke based on middle cerebral artery occlusion for 90 min followed by reperfusion for 7 d, and an in vitro model in primary cultured astrocytes involving oxygen and glucose deprivation for 6 h followed by reoxygenation for 24 h. Both in vivo and in vitro, we found that SB216763, a GSK3β inhibitor, and necrostatin-1 (Nec-1), a RIP1K inhibitor, decreased levels of glial scar markers, including glial fibrillary acidic protein (GFAP), neurocan, and phosphacan. SB216763 and Nec-1 also decreased levels of inflammatory related cytokines, including interleukin-6 (IL-6), interleukin-1 β (IL-1β), and tumor necrosis factor-α (TNF-α). However, only Nec-1 increased the level of interleukin-1 receptor antagonist. Concurrent neutralization of TNF-α, IL-1β, and IL-6 with their antibodies provided better reduction in oxygen and glucose deprivation-induced increases in scar markers than obtained with separate use of each antibody. Further investigations showed that SB216763 reduced the levels of necroptosis-related proteins, including RIP1K, p-RIP1K, RIP3K, p-RIP3K, mixed lineage kinase domain-like protein (MLKL), and p-MLKL, while Nec-1 decreased the expression of p-GSK3β. Compared with Nec-1 (10 μM) and SB216763 (1 μM) alone, Nec-1 and SB216763 in combination reduced levels of GFAP, neurocan, and inflammatory-related cytokines. In conclusion, inhibition of GSK3β or RIP1K reduced glial scar formation induced by ischemic stroke. The underlying mechanisms might be at least, partially related to reducing levels of inflammatory-related cytokines and to blocking an interaction between GSK3β- and RIP1K-mediated pathways.
Collapse
Affiliation(s)
- Jin Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yong-Ming Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yi Guo
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Liang Lin
- Department of Anesthesiology, Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhan-Xiang Wang
- Department of Anesthesiology, Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Feng Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Xin-Yi Dong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Ming Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yi-Fan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Hui-Ling Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
29
|
Triad3A displays a critical role in suppression of cerebral ischemic/reperfusion (I/R) injury by regulating necroptosis. Biomed Pharmacother 2020; 128:110045. [PMID: 32460187 DOI: 10.1016/j.biopha.2020.110045] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/21/2019] [Accepted: 12/26/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemic stroke is a major cause of death and disability worldwide. Necroptosis is known as a form of cell death, playing an essential role in regulating ischemia-induced brain injury. Triad3A is a ubiquitin ligase of the RING-in-between-RING family, and regulates necroptotic cell death under different pathological conditions, including neurodegenerative disorders. In the present study, the effects of Triad3A on experimental stroke were explored on a mouse model with middle cerebral artery occlusion (MCAO). The results indicated that Triad3A expression was markedly induced in the ischemic brain after MCAO operation. The neurons and microglia cells were the major cellular sources for Triad3A induction. Triad3A knockdown enhanced the infarction area, cell death, microglia activity, and the expression levels of pro-inflammatory markers including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, inducible nitric oxide synthase (iNOS), CD32 and CD68 in MCAO mice. Triad3A and necroptosis were triggered in mouse microglia cells treated with oxygen and glucose deprivation (OGD), and in TNFα-incubated mouse hippocampal neuronal cells treated with Z-VAD-fmk, known as a pan-caspase inhibitor. Moreover, Triad3A knockdown accelerated cell death in microglial cells and neurons under these stresses. Furthermore, pre-treatment with necroptosis inhibitor markedly inhibited the cell death promoted by Triad3A silence in brain of mice with MCAO operation, demonstrating that Triad3A could regulate necroptosis to meditate the progression of cerebral I/R injury. Collectively, these finding illustrated that Triad3A could be served as a potential target for stroke therapy.
Collapse
|
30
|
Lin DQ, Cai XY, Wang CH, Yang B, Liang RS. Optimal concentration of necrostatin-1 for protecting against hippocampal neuronal damage in mice with status epilepticus. Neural Regen Res 2020; 15:936-943. [PMID: 31719260 PMCID: PMC6990772 DOI: 10.4103/1673-5374.268903] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/10/2019] [Accepted: 08/13/2019] [Indexed: 02/05/2023] Open
Abstract
Hippocampal neurons undergo various forms of cell death after status epilepticus. Necrostatin-1 specifically inhibits necroptosis mediated by receptor interacting protein kinase 1 (RIP1) and RIP3 receptors. However, there are no reports of necroptosis in mouse models of status epilepticus. Therefore, in this study, we investigated the effects of necrostatin-1 on hippocampal neurons in mice with status epilepticus, and, furthermore, we tested different amounts of the compound to identify the optimal concentration for inhibiting necroptosis and apoptosis. A mouse model of status epilepticus was produced by intraperitoneal injection of kainic acid, 12 mg/kg. Different concentrations of necrostatin-1 (10, 20, 40, and 80 μM) were administered into the lateral ventricle 15 minutes before kainic acid injection. Hippocampal damage was assessed by hematoxylin-eosin staining 24 hours after the model was successfully produced. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining, western blot assay and immunohistochemistry were used to evaluate the expression of apoptosis-related and necroptosis-related proteins. Necrostatin-1 alleviated damage to hippocampal tissue in the mouse model of epilepsy. The 40 μM concentration of necrostatin-1 significantly decreased the number of apoptotic cells in the hippocampal CA1 region. Furthermore, necrostatin-1 significantly downregulated necroptosis-related proteins (MLKL, RIP1, and RIP3) and apoptosis-related proteins (cleaved-Caspase-3, Bax), and it upregulated the expression of anti-apoptotic protein Bcl-2. Taken together, our findings show that necrostatin-1 effectively inhibits necroptosis and apoptosis in mice with status epilepticus, with the 40 μM concentration of the compound having an optimal effect. The experiments were approved by the Animal Ethics Committee of Fujian Medical University, China (approval No. 2016-032) on November 9, 2016.
Collapse
Affiliation(s)
- Dong-Qi Lin
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xin-Ying Cai
- Clinical Research Center, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong Province, China
| | - Chun-Hua Wang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Bin Yang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
- Correspondence to: Ri-Sheng Liang, .
| |
Collapse
|
31
|
Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl Stroke Res 2020; 11:1185-1202. [PMID: 32219729 DOI: 10.1007/s12975-020-00806-z] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is one of the significant causes of morbidity and mortality, affecting millions of people across the globe. Cell injury in the infarct region is an inevitable consequence of focal cerebral ischemia. Subsequent reperfusion exacerbates the harmful effect and increases the infarct volume. These cellular injuries follow either a regulated pathway involving tightly structured signaling cascades and molecularly defined effector mechanisms or a non-regulated pathway, also known as accidental cell death, where the process is biologically uncontrolled. Classical cell death pathways are long established and well reported in several articles that majorly define apoptotic cell death. A recent focus on cell death study also considers investigation on non-classical pathways that are tightly regulated, may or may not involve caspases, but non-apoptotic. Pathological cell death is a cardinal feature of different neurodegenerative diseases. Although ischemia cannot be classified as a neurodegenerative disease, it is a cerebrovascular event where the infarct region exhibits aberrant cell death. Over the past few decades, several therapeutic options have been implicated for ischemic stroke. However, their use has been hampered owing to the number of limitations that they possess. Ischemic penumbral neurons undergo apoptosis and become dysfunctional; however, they are salvageable. Thus, understanding the role of different cell death pathways is crucial to aid in the modern treatment of protecting apoptotic neurons.
Collapse
|
32
|
Heib M, Rose-John S, Adam D. Necroptosis, ADAM proteases and intestinal (dys)function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 353:83-152. [PMID: 32381179 DOI: 10.1016/bs.ircmb.2020.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Recently, an unexpected connection between necroptosis and members of the a disintegrin and metalloproteinase (ADAM) protease family has been reported. Necroptosis represents an important cell death routine which helps to protect from viral, bacterial, fungal and parasitic infections, maintains adult T cell homeostasis and contributes to the elimination of potentially defective organisms before parturition. Equally important for organismal homeostasis, ADAM proteases control cellular processes such as development and differentiation, immune responses or tissue regeneration. Notably, necroptosis as well as ADAM proteases have been implicated in the control of inflammatory responses in the intestine. In this review, we therefore provide an overview of the physiology and pathophysiology of necroptosis, ADAM proteases and intestinal (dys)function, discuss the contribution of necroptosis and ADAMs to intestinal (dys)function, and review the current knowledge on the role of ADAMs in necroptotic signaling.
Collapse
Affiliation(s)
- Michelle Heib
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
33
|
Jantas D, Chwastek J, Grygier B, Lasoń W. Neuroprotective Effects of Necrostatin-1 Against Oxidative Stress-Induced Cell Damage: an Involvement of Cathepsin D Inhibition. Neurotox Res 2020; 37:525-542. [PMID: 31960265 PMCID: PMC7062871 DOI: 10.1007/s12640-020-00164-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
Necroptosis, a recently discovered form of non-apoptotic programmed cell death, can be implicated in many pathological conditions including neuronal cell death. Moreover, an inhibition of this process by necrostatin-1 (Nec-1) has been shown to be neuroprotective in in vitro and in vivo models of cerebral ischemia. However, the involvement of this type of cell death in oxidative stress–induced neuronal cell damage is less recognized. Therefore, we tested the effects of Nec-1, an inhibitor of necroptosis, in the model of hydrogen peroxide (H2O2)-induced cell damage in human neuroblastoma SH-SY5Y and murine hippocampal HT-22 cell lines. The data showed that Nec-1 (10–40 μM) attenuated the cell death induced by H2O2 in undifferentiated (UN-) and neuronal differentiated (RA-) SH-SY5Y cells with a higher efficacy in the former cell type. Moreover, Nec-1 partially reduced cell damage induced by 6-hydroxydopamine in UN- and RA-SH-SY5Y cells. The protective effect of Nec-1 was of similar magnitude as the effect of a caspase-3 inhibitor in both cell phenotypes and this effect were not potentiated after combined treatment. Furthermore, the non-specific apoptosis and necroptosis inhibitor curcumin augmented the beneficial effect of Nec-1 against H2O2-evoked cell damage albeit only in RA-SH-SY5Y cells. Next, it was found that the mechanisms of neuroprotective effect of Nec-1 against H2O2-induced cell damage in SH-SY5Y cells involved the inhibition of lysosomal protease, cathepsin D, but not caspase-3 or calpain activities. In HT-22 cells, Nec-1 was protective in two models of oxidative stress (H2O2 and glutamate) and that effect was blocked by a caspase inhibitor. Our data showed neuroprotective effects of the necroptosis inhibitor, Nec-1, against oxidative stress–induced cell damage and pointed to involvement of cathepsin D inhibition in the mechanism of its action. Moreover, a cell type–specific interplay between necroptosis and apoptosis has been demonstrated.
Collapse
Affiliation(s)
- Danuta Jantas
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| | - Jakub Chwastek
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.,Department of Neurochemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - Beata Grygier
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.,Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7 Street, 30-387, Kraków, Poland
| | - Władysław Lasoń
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| |
Collapse
|
34
|
Guo LM, Wang Z, Li SP, Wang M, Yan WT, Liu FX, Wang CD, Zhang XD, Chen D, Yan J, Xiong K. RIP3/MLKL-mediated neuronal necroptosis induced by methamphetamine at 39°C. Neural Regen Res 2020; 15:865-874. [PMID: 31719251 PMCID: PMC6990769 DOI: 10.4103/1673-5374.268902] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Methamphetamine is one of the most prevalent drugs abused in the world. Methamphetamine abusers usually present with hyperpyrexia (39°C), hallucination and other psychiatric symptoms. However, the detailed mechanism underlying its neurotoxic action remains elusive. This study investigated the effects of methamphetamine + 39°C on primary cortical neurons from the cortex of embryonic Sprague-Dawley rats. Primary cortex neurons were exposed to 1 mM methamphetamine + 39°C. Propidium iodide staining and lactate dehydrogenase release detection showed that methamphetamine + 39°C triggered obvious necrosis-like death in cultured primary cortical neurons, which could be partially inhibited by receptor-interacting protein-1 (RIP1) inhibitor Necrostatin-1 partially. Western blot assay results showed that there were increases in the expressions of receptor-interacting protein-3 (RIP3) and mixed lineage kinase domain-like protein (MLKL) in the primary cortical neurons treated with 1 mM methamphetamine + 39°C for 3 hours. After pre-treatment with RIP3 inhibitor GSK’872, propidium iodide staining and lactate dehydrogenase release detection showed that neuronal necrosis rate was significantly decreased; RIP3 and MLKL protein expression significantly decreased. Immunohistochemistry staining results also showed that the expressions of RIP3 and MLKL were up-regulated in brain specimens from humans who had died of methamphetamine abuse. Taken together, the above results suggest that methamphetamine + 39°C can induce RIP3/MLKL regulated necroptosis, thereby resulting in neurotoxicity. The study protocol was approved by the Medical Ethics Committee of the Third Xiangya Hospital of Central South University, China (approval numbers: 2017-S026 and 2017-S033) on March 7, 2017.
Collapse
Affiliation(s)
- Li-Min Guo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Zhen Wang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province; Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Shi-Ping Li
- Department of Neurology, People's Hospital of Lianhua, Pingxiang, Jiangxi Province, China
| | - Mi Wang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Wei-Tao Yan
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Feng-Xia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Chu-Dong Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Xu-Dong Zhang
- Narcotics Division, Municipal Security Bureau, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
35
|
Sugaya T, Kanno H, Matsuda M, Handa K, Tateda S, Murakami T, Ozawa H, Itoi E. B-RAF V600E Inhibitor Dabrafenib Attenuates RIPK3-Mediated Necroptosis and Promotes Functional Recovery after Spinal Cord Injury. Cells 2019; 8:cells8121582. [PMID: 31817643 PMCID: PMC6953123 DOI: 10.3390/cells8121582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/24/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
The receptor-interacting protein kinase 3 (RIPK3) is a key regulator of necroptosis and is involved in various pathologies of human diseases. We previously reported that RIPK3 expression is upregulated in various neural cells at the lesions and necroptosis contributed to secondary neural tissue damage after spinal cord injury (SCI). Interestingly, recent studies have shown that the B-RAFV600E inhibitor dabrafenib has a function to selectively inhibit RIPK3 and prevents necroptosis in various disease models. In the present study, using a mouse model of thoracic spinal cord contusion injury, we demonstrate that dabrafenib administration in the acute phase significantly inhibites RIPK3-mediated necroptosis in the injured spinal cord. The administration of dabrafenib attenuated secondary neural tissue damage, such as demyelination, neuronal loss, and axonal damage, following SCI. Importantly, the neuroprotective effect of dabrafenib dramatically improved the recovery of locomotor and sensory functions after SCI. Furthermore, the electrophysiological assessment of the injured spinal cord objectively confirmed that the functional recovery was enhanced by dabrafenib. These findings suggest that the B-RAFV600E inhibitor dabrafenib attenuates RIPK3-mediated necroptosis to provide a neuroprotective effect and promotes functional recovery after SCI. The administration of dabrafenib may be a novel therapeutic strategy for treating patients with SCI in the future.
Collapse
Affiliation(s)
- Takehiro Sugaya
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| | - Haruo Kanno
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
- Correspondence: ; Tel.: +81-22-717-7245
| | - Michiharu Matsuda
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| | - Kyoichi Handa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| | - Satoshi Tateda
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| | - Taishi Murakami
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| | - Hiroshi Ozawa
- Department of Orthopaedic Surgery, Tohoku Medical and Pharmaceutical University, Faculty of Medicine, 1-15-1, Fukumuro Miyagino-ku, Sendai 983-8536, Japan;
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (T.S.); (M.M.); (K.H.); (S.T.); (T.M.); (E.I.)
| |
Collapse
|
36
|
Li X, Cheng S, Hu H, Zhang X, Xu J, Wang R, Zhang P. Progranulin protects against cerebral ischemia-reperfusion (I/R) injury by inhibiting necroptosis and oxidative stress. Biochem Biophys Res Commun 2019; 521:569-576. [PMID: 31679689 DOI: 10.1016/j.bbrc.2019.09.111] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
Ischemic stroke is a leading cause of mortality and disability worldwide. Nevertheless, its molecular mechanisms have not yet been adequately illustrated. Progranulin (PGRN) is a secreted glycoprotein with pleiotropic functions. In the present study, we found that PGRN expression was markedly reduced in mice after stroke onset through middle cerebral artery occlusion (MCAO). We also showed that necroptosis was a mechanism underlying cerebral I/R injury. Importantly, PGRN knockdown in vivo significantly promoted the infarction volume and neurological deficits scores in mice after MCAO surgery. Necroptosis induced by MCAO was further accelerated by PGRN knockdown, as evidenced by the promoted expression of phosphorylated receptor-interacting protein (RIP) 1 kinase (RIPK1), RIPK3 and mixed lineage kinase domain-like (MLKL), which was accompanied with increased expression of cleaved Caspase-8 and Caspase-3. However, PGRN over-expression was neuroprotective. Additionally, PGRN-regulated ischemic stroke was related to ROS accumulation that MCAO-mice with PGRN knockdown exhibited severe oxidative stress, as proved by the aggravated malondialdehyde (MDA) and lipid peroxidation (LPO) contents, and the decreased superoxide dismutase (SOD) activity. However, PGRN over-expression in mice with cerebral ischemia showed anti-oxidative effects. Finally, PGRN was found to attenuate oxidative damage partly via its regulatory effects on necroptosis. Therefore, promoting PGRN expression could reduced cerebral I/R-induced brain injury by suppressing neroptosis and associated reactive oxygen species (ROS) production. These data elucidated that PGRN might provide an effective therapeutic treatment for ischemic stroke.
Collapse
Affiliation(s)
- Xiaogang Li
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shaoli Cheng
- Basic Medical Experimental Teaching Center, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Basic Medical Experimental Teaching Center, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaotian Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Jiehua Xu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Rui Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Pengbo Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
37
|
Gurunathan S, Jeyaraj M, Kang MH, Kim JH. Mitochondrial Peptide Humanin Protects Silver Nanoparticles-Induced Neurotoxicity in Human Neuroblastoma Cancer Cells (SH-SY5Y). Int J Mol Sci 2019; 20:ijms20184439. [PMID: 31505887 PMCID: PMC6770400 DOI: 10.3390/ijms20184439] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
The extensive usage of silver nanoparticles (AgNPs) as medical products such as antimicrobial and anticancer agents has raised concerns about their harmful effects on human beings. AgNPs can potentially induce oxidative stress and apoptosis in cells. However, humanin (HN) is a small secreted peptide that has cytoprotective and neuroprotective cellular effects. The aim of this study was to assess the harmful effects of AgNPs on human neuroblastoma SH-SY5Y cells and also to investigate the protective effect of HN from AgNPs-induced cell death, mitochondrial dysfunctions, DNA damage, and apoptosis. AgNPs were prepared with an average size of 18 nm diameter to study their interaction with SH-SY5Y cells. AgNPs caused a dose-dependent decrease of cell viability and proliferation, induced loss of plasma-membrane integrity, oxidative stress, loss of mitochondrial membrane potential (MMP), and loss of ATP content, amongst other effects. Pretreatment or co-treatment of HN with AgNPs protected cells from several of these AgNPs induced adverse effects. Thus, this study demonstrated for the first time that HN protected neuroblastoma cells against AgNPs-induced neurotoxicity. The mechanisms of the HN-mediated protective effect on neuroblastoma cells may provide further insights for the development of novel therapeutic agents against neurodegenerative diseases.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Muniyandi Jeyaraj
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
38
|
Deng XX, Li SS, Sun FY. Necrostatin-1 Prevents Necroptosis in Brains after Ischemic Stroke via Inhibition of RIPK1-Mediated RIPK3/MLKL Signaling. Aging Dis 2019; 10:807-817. [PMID: 31440386 PMCID: PMC6675533 DOI: 10.14336/ad.2018.0728] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/28/2018] [Indexed: 12/11/2022] Open
Abstract
Pharmacological studies have indirectly shown that necroptosis participates in ischemic neuronal death. However, its mechanism has yet to be elucidated in the ischemic brain. TNFα-triggered RIPK1 kinase activation could initiate RIPK3/MLKL-mediated necroptosis under inhibition of caspase-8. In the present study, we performed middle cerebral artery occlusion (MCAO) to induce cerebral ischemia in rats and used immunoblotting and immunostaining combined with pharmacological analysis to study the mechanism of necroptosis in ischemic brains. In the ipsilateral hemisphere, we found that ischemia induced the increase of (i) RIPK1 phosphorylation at the Ser166 residue (p-RIPK1), representing active RIPK1 kinase and (ii) the number of cells that were double stained with P-RIPK1 (Ser166) (p-RIPK1+) and TUNEL, a label of DNA double-strand breaks, indicating cell death. Furthermore, ischemia induced activation of downstream signaling factors of RIPK1, RIPK3 and MLKL, as well as the formation of mature interleukin-1β (IL-1β). Treatment with necrostatin-1 (Nec-1), an inhibitor of necroptosis, significantly decreased ischemia-induced increase of p-RIPK1 expression and p-RIPK1+ neurons, which showed protection from brain damage. Meanwhile, Nec-1 reduced RIPK3, MLKL and p-MLKL expression levels and mature IL-1β formation in Nec-1 treated ischemic brains. Our results clearly demonstrated that phosphorylation of RIPK1 at the Ser166 residue was involved in the pathogenesis of necroptosis in the brains after ischemic injury. Nec-1 treatment protected brains against ischemic necroptosis by reducing the activation of RIPK1 and inhibiting its downstream signaling pathways. These results provide direct in vivo evidence that phosphorylated RIPK1 (Ser 166) plays an important role in the initiation of RIPK3/MLKL-dependent necroptosis in the pathogenesis of ischemic stroke in the rodent brain.
Collapse
Affiliation(s)
- Xu-Xu Deng
- 1Department of Neurobiology, School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,2Institute for Basic Research on Aging and Medicine, the State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shan-Shan Li
- 1Department of Neurobiology, School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,2Institute for Basic Research on Aging and Medicine, the State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng-Yan Sun
- 1Department of Neurobiology, School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,2Institute for Basic Research on Aging and Medicine, the State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,3Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Hribljan V, Lisjak D, Petrović DJ, Mitrečić D. Necroptosis is one of the modalities of cell death accompanying ischemic brain stroke: from pathogenesis to therapeutic possibilities. Croat Med J 2019. [PMID: 31044583 PMCID: PMC6509625 DOI: 10.3325/cmj.2019.60.121] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Due to very limited therapeutic options, ischemic brain injury is one of the leading causes of death and lifelong disability worldwide, which imposes enormous public health burden. One of the main events occurring with ischemic brain stroke is cell death. Necroptosis is a type of cell death described as a regulated necrosis characterized by cell membrane disruption mediated by phosphorylated mixed lineage kinase like protein (MLKL). It can be triggered by activation of death receptors (eg, FAS, TNFR1), which lead to receptor-interacting serine/threonine-protein kinase 3 (RIPK3) activation by RIPK1 in the absence of active caspase-8. Here, we review articles that have reported that necroptosis significantly contributes to negative events occurring with the ischemic brain stroke, and that its inhibition is protective both in vitro and in vivo. We also review articles describing positive effects obtained by reducing necroptosis, including the reduction of infarct volume and improved functional recovery in animal models. Since necroptosis is characterized by cell content leakage and subsequent inflammation, in addition to reducing cell death, inhibition of necroptosis in ischemic brain stroke also reduces some inflammatory cytokines. By comparing various approaches in inhibition of necroptosis, we analyze the achieved effects from the perspective of controlling necroptosis as a part of future therapeutic interventions in brain ischemia.
Collapse
Affiliation(s)
- Valentina Hribljan
- Valentina Hribljan, Laboratory for Stem Cells, Department for Neurogenetics, Medical Genetics and Regenerative Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, HR-10000 Zagreb, Croatia,
| | | | | | | |
Collapse
|
40
|
Han F, Guan X, Guo W, Lu B. Therapeutic potential of a TrkB agonistic antibody for ischemic brain injury. Neurobiol Dis 2019; 127:570-581. [DOI: 10.1016/j.nbd.2019.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022] Open
|
41
|
Shen Y, Peng C, Bai Q, Ding Y, Yi X, Du H, He L, Zhou D, Chen X. Epigenome-Wide Association Study Indicates Hypomethylation of MTRNR2L8 in Large-Artery Atherosclerosis Stroke. Stroke 2019; 50:1330-1338. [PMID: 31084332 DOI: 10.1161/strokeaha.118.023436] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Ischemic stroke, a complex and heterogeneous disease, is the second leading cause of death worldwide. Genetic factors and epigenetic modification contribute to the pathogenesis of this disease. However, the effects of epigenetic factors on this disease have not been systematically investigated. Our study was designed to identify methylation alterations in large-artery atherosclerotic stroke. Methods- We conducted an epigenome-wide association analysis of large-artery atherosclerotic stroke using an Infinium HumanMethylation450 array (cases:controls=12:12), and the differentially methylated loci were validated in 2 cohorts (cases:controls, 110:122 and 191:191, respectively) using a Sequenom EpiTYPER assay. Results- In the screening stage, 1012 differentially methylated CpG sites annotated in 672 genes were found to be significantly associated with large-artery atherosclerotic stroke (mean methylation difference >5%, P<0.01). Disease, Gene Ontology, and pathway analysis highlighted the enrichment of these differentially methylated genes in cardiovascular, metabolic, neurological and immune-related functional gene clusters ( P<0.05). We identified a differentially methylated region in the promoter of a humanin gene ( MTRNR2L8, mean methylation difference=-13.01%, P=8.86×10-14). We constructed a diagnostic prediction model that was based on the mean number of significantly changed CpG loci in MTRNR2L8 and showed high diagnostic specificity and sensitivity ( P<0.0001, area under the curve=0.774). Conclusions- Together, these findings demonstrate that DNA methylation plays an important role in large-artery atherosclerotic stroke and that methylation of MTRNR2L8 is a potential therapeutic target and diagnostic biomarker for stroke.
Collapse
Affiliation(s)
- Yupei Shen
- From the Department of Neurology, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine (Y.S., C.P., Y.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) (Y.S., X.Y., H.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai (Y.S., D.Z., X.C.)
| | - Chen Peng
- From the Department of Neurology, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine (Y.S., C.P., Y.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Department of Neurology, Shanghai Eighth People's Hospital affiliated with Jiangsu University, China (C.P., Y.D.)
| | - Qingke Bai
- Departments of Neurology, Pudong People's Hospital, Shanghai, China (Q.B.)
| | - Ying Ding
- From the Department of Neurology, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine (Y.S., C.P., Y.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Department of Neurology, Shanghai Eighth People's Hospital affiliated with Jiangsu University, China (C.P., Y.D.)
| | - Xin Yi
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) (Y.S., X.Y., H.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
| | - Huihui Du
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) (Y.S., X.Y., H.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
| | - Lin He
- From the Department of Neurology, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine (Y.S., C.P., Y.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) (Y.S., X.Y., H.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
| | - Daizhan Zhou
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai (Y.S., D.Z., X.C.)
- Institute of Medical Genetics, Tongji University, Shanghai, China (D.Z.)
| | - Xu Chen
- From the Department of Neurology, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine (Y.S., C.P., Y.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) (Y.S., X.Y., H.D., L.H., X.C.), Shanghai Jiao Tong University, PR China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai (Y.S., D.Z., X.C.)
| |
Collapse
|
42
|
Hribljan V, Lisjak D, Petrović DJ, Mitrečić D. Necroptosis is one of the modalities of cell death accompanying ischemic brain stroke: from pathogenesis to therapeutic possibilities. Croat Med J 2019; 60:121-126. [PMID: 31044583 PMCID: PMC6509625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/12/2019] [Indexed: 10/12/2023] Open
Abstract
Due to very limited therapeutic options, ischemic brain injury is one of the leading causes of death and lifelong disability worldwide, which imposes enormous public health burden. One of the main events occurring with ischemic brain stroke is cell death. Necroptosis is a type of cell death described as a regulated necrosis characterized by cell membrane disruption mediated by phosphorylated mixed lineage kinase like protein (MLKL). It can be triggered by activation of death receptors (eg, FAS, TNFR1), which lead to receptor-interacting serine/threonine-protein kinase 3 (RIPK3) activation by RIPK1 in the absence of active caspase-8. Here, we review articles that have reported that necroptosis significantly contributes to negative events occurring with the ischemic brain stroke, and that its inhibition is protective both in vitro and in vivo. We also review articles describing positive effects obtained by reducing necroptosis, including the reduction of infarct volume and improved functional recovery in animal models. Since necroptosis is characterized by cell content leakage and subsequent inflammation, in addition to reducing cell death, inhibition of necroptosis in ischemic brain stroke also reduces some inflammatory cytokines. By comparing various approaches in inhibition of necroptosis, we analyze the achieved effects from the perspective of controlling necroptosis as a part of future therapeutic interventions in brain ischemia.
Collapse
Affiliation(s)
- Valentina Hribljan
- Valentina Hribljan, Laboratory for Stem Cells, Department for Neurogenetics, Medical Genetics and Regenerative Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, HR-10000 Zagreb, Croatia,
| | | | | | | |
Collapse
|
43
|
TRAF2 protects against cerebral ischemia-induced brain injury by suppressing necroptosis. Cell Death Dis 2019; 10:328. [PMID: 30988281 PMCID: PMC6465397 DOI: 10.1038/s41419-019-1558-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022]
Abstract
Necroptosis contributes to ischemia-induced brain injury. Tumor necrosis factor (TNF) receptor associated factor 2 (TRAF2) has been reported to suppress necroptotic cell death under several pathological conditions. In this study, we investigated the role of TRAF2 in experimental stroke using a mouse middle cerebral artery occlusion (MCAO) model and in vitro cellular models. TRAF2 expression in the ischemic brain was assessed with western blot and real-time RT-PCR. Gene knockdown of TRAF2 by lentivirus was utilized to investigate the role of TRAF2 in stroke outcomes. The expression of TRAF2 was significantly induced in the ischemic brain at 24 h after reperfusion, and neurons and microglia were two of the cellular sources of TRAF2 induction. Striatal knockdown of TRAF2 increased infarction size, cell death, microglial activation and the expression of pro-inflammatory markers at 24 h after reperfusion. TRAF2 expression and necroptosis were induced in mouse primary microglia treated with conditioned medium collected from neurons subject to oxygen and glucose deprivation (OGD) and in TNFα-treated mouse hippocampal neuronal HT-22 cells in the presence of the pan-caspase inhibitor Z-VAD. In addition, TRAF2 knockdown exacerbated microglial cell death and neuronal cell death under these conditions. Moreover, pre-treatment with a specific necroptosis inhibitor necrostatin-1 (nec-1) suppressed the cell death exacerbated by TRAF2 knockdown in the brain following MCAO, indicating that TRAF2 impacted ischemic brain damage through necroptosis mechanism. Taken together, our results demonstrate that TRAF2 is a novel regulator of cerebral ischemic injury.
Collapse
|
44
|
Magtanong L, Dixon SJ. Ferroptosis and Brain Injury. Dev Neurosci 2019; 40:382-395. [PMID: 30820017 PMCID: PMC6658337 DOI: 10.1159/000496922] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/14/2019] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by the iron-dependent accumulation of toxic lipid reactive oxygen species. Small-molecule screening and subsequent optimization have yielded potent and specific activators and inhibitors of this process. These compounds have been employed to dissect the lethal mechanism and implicate this process in pathological cell death events observed in many tissues, including the brain. Indeed, ferroptosis is emerging as an important mechanism of cell death during stroke, intracerebral hemorrhage, and other acute brain injuries, and may also play a role in certain degenerative brain disorders. Outstanding issues include the practical need to identify molecular markers of ferroptosis that can be used to detect and study this process in vivo, and the more basic problem of understanding the relationship between ferroptosis and other forms of cell death that can be triggered in the brain during injury.
Collapse
Affiliation(s)
- Leslie Magtanong
- Department of Biology, Stanford University, Stanford, California, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, California, USA,
| |
Collapse
|
45
|
Jun-Long H, Yi L, Bao-Lian Z, Jia-Si L, Ning Z, Zhou-Heng Y, Xue-Jun S, Wen-Wu L. Necroptosis Signaling Pathways in Stroke: From Mechanisms to Therapies. Curr Neuropharmacol 2018; 16:1327-1339. [PMID: 29663889 PMCID: PMC6251040 DOI: 10.2174/1570159x16666180416152243] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 11/20/2017] [Accepted: 02/28/2018] [Indexed: 12/17/2022] Open
Abstract
It has been confirmed that apoptosis, autophagy and necrosis are the three major modes of cell death. For a long time, necrosis is regarded as a deranged or accidental cell demise. In recent years, there is evidence showing that necrotic cell death can be a well regulated and orchestrated event, which is also known as programmed cell death or “necroptosis”. Necroptosis can be triggered by a variety of external stimuli and regulated by a caspase-independent pathway. It plays a key role in the pathogenesis of some diseases including neurological diseases. In the past two decades, a variety of studies have revealed that the necroptosis related pathway is activated in stroke, and plays a crucial role in the pathogenesis of stroke. Moreover, necroptosis may serve as a potential target in the therapy of stroke because genetic or pharmacological inhibition of necroptosis has been shown to be neuroprotective in stroke in vitro and in vivo. In this review, we briefly summarize re-cent advances in necroptosis, introduce the mechanism and strategies targeting necroptosis in stroke, and finally propose some issues in the treatment of stroke by targeting necroptosis
Collapse
Affiliation(s)
- Huang Jun-Long
- Department of Navy Aviation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Li Yi
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Zhao Bao-Lian
- Department of Naval Clinical Medicine, Second Military Medical University, Shanghai 200433, China
| | - Li Jia-Si
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhang Ning
- Department of Navy Aviation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Ye Zhou-Heng
- Department of Navy Aviation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Sun Xue-Jun
- Department of Navy Aviation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Liu Wen-Wu
- Department of Diving and Hyperbaric Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
46
|
Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Bøtker HE, Carr RD, Cunningham J, Ghosh AK, Heusch G, Ibanez B, Kleinbongard P, Lecour S, Maddock H, Ovize M, Walker M, Wiart M, Yellon DM. The 10th Biennial Hatter Cardiovascular Institute workshop: cellular protection-evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol 2018; 113:43. [PMID: 30310998 PMCID: PMC6182684 DOI: 10.1007/s00395-018-0704-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
Due to its poor capacity for regeneration, the heart is particularly sensitive to the loss of contractile cardiomyocytes. The onslaught of damage caused by ischaemia and reperfusion, occurring during an acute myocardial infarction and the subsequent reperfusion therapy, can wipe out upwards of a billion cardiomyocytes. A similar program of cell death can cause the irreversible loss of neurons in ischaemic stroke. Similar pathways of lethal cell injury can contribute to other pathologies such as left ventricular dysfunction and heart failure caused by cancer therapy. Consequently, strategies designed to protect the heart from lethal cell injury have the potential to be applicable across all three pathologies. The investigators meeting at the 10th Hatter Cardiovascular Institute workshop examined the parallels between ST-segment elevation myocardial infarction (STEMI), ischaemic stroke, and other pathologies that cause the loss of cardiomyocytes including cancer therapeutic cardiotoxicity. They examined the prospects for protection by remote ischaemic conditioning (RIC) in each scenario, and evaluated impasses and novel opportunities for cellular protection, with the future landscape for RIC in the clinical setting to be determined by the outcome of the large ERIC-PPCI/CONDI2 study. It was agreed that the way forward must include measures to improve experimental methodologies, such that they better reflect the clinical scenario and to judiciously select combinations of therapies targeting specific pathways of cellular death and injury.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sapna Arjun
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Maryna V Basalay
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Robert M Bell
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Richard D Carr
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- MSD A/S, Copenhagen, Denmark
| | - John Cunningham
- Centre for Nephrology, UCL Medical School, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Arjun K Ghosh
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Gerd Heusch
- West German Heart and Vascular Center, Institute for Pathophysiology, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades CardioVasculares, Madrid, Spain
- IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Petra Kleinbongard
- West German Heart and Vascular Center, Institute for Pathophysiology, University of Essen Medical School, Essen, Germany
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Helen Maddock
- Centre for Sport, Exercise and Life Sciences, Faculty of Health and Life Sciences, Coventry University, Priory Street, Coventry, CV1 5FB, UK
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Service d'explorations Fonctionnelles Cardiovasculaires Groupement Hospitalier Est, 59 Boulevard Pinel, 69500, Bron, France
| | - Malcolm Walker
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Marlene Wiart
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Service d'explorations Fonctionnelles Cardiovasculaires Groupement Hospitalier Est, 59 Boulevard Pinel, 69500, Bron, France
- CNRS, Lyon, France
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
47
|
Zhan L, Lu Z, Zhu X, Xu W, Li L, Li X, Chen S, Sun W, Xu E. Hypoxic preconditioning attenuates necroptotic neuronal death induced by global cerebral ischemia via Drp1-dependent signaling pathway mediated by CaMKIIα inactivation in adult rats. FASEB J 2018; 33:1313-1329. [PMID: 30148677 DOI: 10.1096/fj.201800111rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hypoxic preconditioning (HPC) alleviates the selective and delayed neuronal death in the hippocampal CA1 region induced by transient global cerebral ischemia (tGCI). This type of cell death may include different programmed cell death mechanisms, namely, apoptosis and necroptosis. Although apoptotic signaling is well defined, the mechanisms that underlie neuronal necroptosis are yet to be fully elucidated. In this study, we investigated whether HPC protects neurons from cerebral ischemia-induced necroptosis. We observed that tGCI up-regulated the expression of receptor-interacting protein (RIP) 3 and increased the interaction of RIP1-RIP3 in CA1 at the early stage of reperfusion. The pretreatment with HPC or necrostatin-1 decreased the expression of RIP3 and the formation of RIP1-RIP3 after tGCI. We also found that HPC decreased the expression and the activity of caspase-8 in CA1 after tGCI, and notably, the pretreatment with Z-VAD-FMK, a pan-caspase inhibitor, did not trigger necroptosis but attenuated the tGCI-induced neuronal damage. Furthermore, we demonstrated that HPC decreased the activation of calcium-calmodulin kinase (CaMK) IIα and the interaction of RIP1 and CaMKIIα induced by tGCI. Intriguingly, the pretreatment with a CaMKs inhibitor KN-93 before tGCI resulted in significantly reduced RIP1-3 interaction and tGCI-induced neuronal damage. Finally, we ascertained that HPC prevented the dephosphorylation of dynamin-related protein 1 (Drp1)-Ser637 (serine 637) and inhibited the translocation of Drp1 to mitochondria induced by tGCI. Importantly, the treatment with a Drp1 inhibitor Mdivi-1 or necrostatin-1 before tGCI also abolished Drp1 dephosphorylation at Ser637 and mitochondrial translocation. Taken together, our results highlight that HPC attenuates necroptotic neuronal death induced by tGCI via Drp1-dependent mitochondrial signaling pathways mediated by CaMKIIα inactivation.-Zhan, L., Lu, Z., Zhu, X., Xu, W., Li, L., Li, X., Chen, S., Sun, W., Xu, E. Hypoxic preconditioning attenuates necroptotic neuronal death induced by global cerebral ischemia via Drp1-dependent signaling pathway mediated by CaMKIIα inactivation in adult rats.
Collapse
Affiliation(s)
- Lixuan Zhan
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Lu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou Huai Hospital, Guangzhou, China
| | - Xinyong Zhu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wensheng Xu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Luxi Li
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xinyu Li
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Siyuan Chen
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weiwen Sun
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - En Xu
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education, Guangzhou, China.,Institute of Neurosciences, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Neurology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
48
|
Li W, Liu J, Chen JR, Zhu YM, Gao X, Ni Y, Lin B, Li H, Qiao SG, Wang C, Zhang HL, Ao GZ. Neuroprotective Effects of DTIO, A Novel Analog of Nec-1, in Acute and Chronic Stages After Ischemic Stroke. Neuroscience 2018; 390:12-29. [PMID: 30076999 DOI: 10.1016/j.neuroscience.2018.07.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 02/04/2023]
Abstract
Receptor-interacting protein 1 kinase (RIP1K) plays a key role in necroptosis. Necrostatin-1 (Nec-1), a specific inhibitor of RIP1K, provides neuroprotection against ischemic brain injury, associating with inhibition of inflammation. Recently, our group synthesized a novel analog of Nec-1, 5-(3',5'-dimethoxybenzal)-2-thio-imidazole-4-ketone (DTIO). The present study investigated the effect of DTIO on ischemic stroke-induced brain injury in both acute and chronic phase and its underlying mechanism. In vivo, DTIO treatment reduced infarct volume and improved neurological deficits in the acute phase after permanent middle cerebral artery occlusion (pMCAO) and it also attenuated brain atrophy and promoted brain functional recovery in the chronic phase post-cerebral ischemia/reperfusion (I/R). In vitro, DTIO treatment decreased lactate dehydrogenase (LDH) leakage and necrotic cell death in the oxygen and glucose deprivation (OGD) or oxygen and glucose deprivation and reoxygenation (OGD/R)-induced neuronal or astrocytic cell injury. Simultaneously, DTIO suppressed the production and release of inflammatory cytokines, and reduced the formation of glial scar. Homology modeling analysis illustrated that DTIO had an ability of binding to RIP1K. Furthermore, immunoprecipitation analysis showed that DTIO inhibited the phosphorylation of RIP1K and decreased the interaction between the RIP1K and RIP3K. In addition, knockdown of RIP1K had neuroprotective effects and inhibited the release of proinflammatory cytokines, but didn't have a significant effect on DTIO-mediated neuroprotection. In conclusion, DTIO has protective effects on acute ischemic stroke and promotes functional recovery during chronic phase, associating with protecting ischemic neurons and astrocytes, inhibiting inflammation, and lessening the glial scar formation via inhibiting of the RIP1K.
Collapse
Affiliation(s)
- Wei Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jin Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jie-Ru Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yong-Ming Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xue Gao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yong Ni
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huanqiu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shi-Gang Qiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China; Department of Anesthesiology and Perioperative Medicine, Suzhou Science and Technology Town Hospital, and Institute of Clinical Medicine, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215153, China
| | - Chen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China; Department of Anesthesiology and Perioperative Medicine, Suzhou Science and Technology Town Hospital, and Institute of Clinical Medicine, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215153, China
| | - Hui-Ling Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Gui-Zhen Ao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
49
|
Chen Q, Kang J, Fu C. The independence of and associations among apoptosis, autophagy, and necrosis. Signal Transduct Target Ther 2018; 3:18. [PMID: 29967689 PMCID: PMC6026494 DOI: 10.1038/s41392-018-0018-5] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 05/02/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Cell death is an essential biological process for physiological growth and development. Three classical forms of cell death-apoptosis, autophagy, and necrosis-display distinct morphological features by activating specific signaling pathways. With recent research advances, we have started to appreciate that these cell death processes can cross-talk through interconnecting, even overlapping, signaling pathways, and the final cell fate is the result of the interplay of different cell death programs. This review provides an insight into the independence of and associations among these three types of cell death and explores the significance of cell death under the specific conditions of human diseases, particularly neurodegenerative diseases and cancer.
Collapse
Affiliation(s)
- Qi Chen
- 1College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, 310018 China.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018 China
| | - Jian Kang
- 3Cancer Signalling Laboratory, Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, 305 Grattan street, Melbourne, VIC 3000 Australia
| | - Caiyun Fu
- 1College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, 310018 China.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018 China.,4Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, 555 Mission Bay Blvd. South, San Francisco, CA 94158 USA.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014 China
| |
Collapse
|
50
|
Wang Z, Guo LM, Wang Y, Zhou HK, Wang SC, Chen D, Huang JF, Xiong K. Inhibition of HSP90α protects cultured neurons from oxygen-glucose deprivation induced necroptosis by decreasing RIP3 expression. J Cell Physiol 2018; 233:4864-4884. [PMID: 29334122 DOI: 10.1002/jcp.26294] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/14/2017] [Indexed: 01/03/2023]
Abstract
Heat shock protein 90α (HSP90α) maintains cell stabilization and regulates cell death, respectively. Recent studies have shown that HSP90α is involved in receptor interacting protein 3 (RIP3)-mediated necroptosis in HT29 cells. It is known that oxygen and glucose deprivation (OGD) can induce necroptosis, which is regulated by RIP3 in neurons. However, it is still unclear whether HSP90α participates in the process of OGD-induced necroptosis in cultured neurons via the regulation of RIP3. Our study found that necroptosis occurs in primary cultured cortical neurons and PC-12 cells following exposure to OGD insult. Additionally, the expression of RIP3/p-RIP3, MLKL/p-MLKL, and the RIP1/RIP3 complex (necrosome) significantly increased following OGD, as measured through immunofluorescence (IF) staining, Western blotting (WB), and immunoprecipitation (IP) assay. Additionally, data from computer simulations and IP assays showed that HSP90α interacts with RIP3. In addition, HSP90α was overexpressed following OGD in cultured neurons, as measured through WB and IF staining. Inhibition of HSP90α in cultured neurons, using the specific inhibitor, geldanamycin (GA), and siRNA/shRNA of HSP90α, protected cultured neurons from necrosis. Our study showed that the inhibitor of HSP90α, GA, rescued cultured neurons not only by decreasing the expression of total RIP3/MLKL, but also by decreasing the expression of p-RIP3/p-MLKL and the RIP1/RIP3 necrosome. In this study, we reveal that inhibition of HSP90α protects primary cultured cortical neurons and PC-12 cells from OGD-induced necroptosis through the modulation of RIP3 expression.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Li-Min Guo
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Hong-Kang Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Shu-Chao Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ju-Fang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|