1
|
Obeng E, Shen B, Wang W, Xie Z, Zhang W, Li Z, Yao Q, Wu W. Engineered bio-functional material-based nerve guide conduits for optic nerve regeneration: a view from the cellular perspective, challenges and the future outlook. Regen Biomater 2024; 12:rbae133. [PMID: 39776856 PMCID: PMC11703557 DOI: 10.1093/rb/rbae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/16/2024] [Accepted: 11/03/2024] [Indexed: 01/11/2025] Open
Abstract
Nerve injuries can be tantamount to severe impairment, standard treatment such as the use of autograft or surgery comes with complications and confers a shortened relief. The mechanism relevant to the regeneration of the optic nerve seems yet to be fully uncovered. The prevailing rate of vision loss as a result of direct or indirect insult on the optic nerve is alarming. Currently, the use of nerve guide conduits (NGC) to some extent has proven reliable especially in rodents and among the peripheral nervous system, a promising ground for regeneration and functional recovery, however in the optic nerve, this NGC function seems quite unfamous. The insufficient NGC application and the unabridged regeneration of the optic nerve could be a result of the limited information on cellular and molecular activities. This review seeks to tackle two major factors (i) the cellular and molecular activity involved in traumatic optic neuropathy and (ii) the NGC application for the optic nerve regeneration. The understanding of cellular and molecular concepts encompassed, ocular inflammation, extrinsic signaling and intrinsic signaling for axon growth, mobile zinc role, Ca2+ factor associated with the optic nerve, alternative therapies from nanotechnology based on the molecular information and finally the nanotechnological outlook encompassing applicable biomaterials and the use of NGC for regeneration. The challenges and future outlook regarding optic nerve regenerations are also discussed. Upon the many approaches used, the comprehensive role of the cellular and molecular mechanism may set grounds for the efficient application of the NGC for optic nerve regeneration.
Collapse
Affiliation(s)
- Enoch Obeng
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Baoguo Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenyuan Xie
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wenyi Zhang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhixing Li
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Qinqin Yao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang 325000, China
| |
Collapse
|
2
|
Suryavanshi P, Langton R, Fairhead K, Glykys J. Brief and Diverse Excitotoxic Insults Increase the Neuronal Nuclear Membrane Permeability in the Neonatal Brain, Resulting in Neuronal Dysfunction and Cell Death. J Neurosci 2024; 44:e0350242024. [PMID: 39214703 PMCID: PMC11466074 DOI: 10.1523/jneurosci.0350-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Neuronal cytotoxic edema is implicated in neuronal injury and death, yet mitigating brain edema with osmotic and surgical interventions yields poor clinical outcomes. Importantly, neuronal swelling and its downstream consequences during early brain development remain poorly investigated, and new treatment approaches are needed. We explored Ca2+-dependent downstream effects after neuronal cytotoxic edema caused by diverse injuries in mice of both sexes using multiphoton Ca2+ imaging in vivo [Postnatal Day (P)12-17] and in acute brain slices (P8-12). After different excitotoxic insults, cytosolic GCaMP6s translocated into the nucleus after a few minutes in a subpopulation of neurons, persisting for hours. We used an automated morphology-detection algorithm to detect neuronal soma and quantified the nuclear translocation of GCaMP6s as the nuclear to cytosolic intensity (N/C ratio). Elevated neuronal N/C ratios occurred concurrently with persistent elevation in Ca2+ loads and could also occur independently from neuronal swelling. Electron microscopy revealed that the nuclear translocation was associated with the increased nuclear pore size. The nuclear accumulation of GCaMP6s in neurons led to neocortical circuit dysfunction, mitochondrial pathology, and increased cell death. Inhibiting calpains, a family of Ca2+-activated proteases, prevented elevated N/C ratios and neuronal swelling. In summary, in the developing brain, we identified a calpain-dependent alteration of nuclear transport in a subpopulation of neurons after disease-relevant insults leading to long-term circuit dysfunction and cell death. The nuclear translocation of GCaMP6 and other cytosolic proteins after acute excitotoxicity can be an early biomarker of brain injury in the developing brain.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
| | - Rachel Langton
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
| | - Kimberly Fairhead
- Biomedical Sciences, College of Liberal Arts and Sciences, The University of Iowa, Iowa City, Iowa 52242
| | - Joseph Glykys
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
- Department of Neurology, The University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
3
|
Rahi V, Kaundal RK. Exploring the intricacies of calcium dysregulation in ischemic stroke: Insights into neuronal cell death and therapeutic strategies. Life Sci 2024; 347:122651. [PMID: 38642844 DOI: 10.1016/j.lfs.2024.122651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/16/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Calcium ion (Ca2+) dysregulation is one of the main causes of neuronal cell death and brain damage after cerebral ischemia. During ischemic stroke, the ability of neurons to maintain Ca2+ homeostasis is compromised. Ca2+ regulates various functions of the nervous system, including neuronal activity and adenosine triphosphate (ATP) production. Disruptions in Ca2+ homeostasis can trigger a cascade of events, including activation of the unfolded protein response (UPR) pathway, which is associated with endoplasmic reticulum (ER) stress and mitochondrial dysfunction. This response occurs when the cell is unable to manage protein folding within the ER due to various stressors, such as a high influx of Ca2+. Consequently, the UPR is initiated to restore ER function and alleviate stress, but prolonged activation can lead to mitochondrial dysfunction and, ultimately, cell death. Hence, precise regulation of Ca2+ within the cell is mandatory. The ER and mitochondria are two such organelles that maintain intracellular Ca2+ homeostasis through various calcium-operating channels, including ryanodine receptors (RyRs), inositol trisphosphate receptors (IP3Rs), sarco/endoplasmic reticulum calcium ATPases (SERCAs), the mitochondrial Na+/Ca2+ exchanger (NCLX), the mitochondrial calcium uniporter (MCU) and voltage-dependent anion channels (VDACs). These channels utilize Ca2+ sequestering and release mechanisms to maintain intracellular Ca2+ homeostasis and ensure proper cellular function and survival. The present review critically evaluates the significance of Ca2+ and its physiological role in cerebral ischemia. We have compiled recent findings on calcium's role and emerging treatment strategies, particularly targeting mitochondria and the endoplasmic reticulum, to address Ca2+ overload in cerebral ischemia.
Collapse
Affiliation(s)
- Vikrant Rahi
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226 002, India
| | - Ravinder K Kaundal
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226 002, India.
| |
Collapse
|
4
|
Wang P, Ouyang J, Zhou K, Hu D, Zhang S, Zhang A, Yang Y. Olesoxime protects against cisplatin-induced acute kidney injury by attenuating mitochondrial dysfunction. Biomed J 2024; 48:100730. [PMID: 38643825 PMCID: PMC11751417 DOI: 10.1016/j.bj.2024.100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction is a critical factor in the pathogenesis of acute kidney injury (AKI). Agents that ameliorate mitochondrial dysfunction hold potential for AKI treatment. The objective of this study was to investigate the impact of olesoxime, a novel mitochondrial-targeted agent, on cisplatin-induced AKI. METHODS In vivo, a cisplatin-induced AKI mouse model was established by administering a single intraperitoneal dose of cisplatin (25 mg/kg) to male C57BL/6 mice for 72 hours, followed by gavage of either olesoxime or a control solution. In vitro, human proximal tubular HK2 cells were cultured and subjected to treatments with cisplatin, either in the presence or absence of olesoxime. RESULTS In vivo, our findings demonstrated that olesoxime administration significantly mitigated the nephrotoxic effects of cisplatin in mice, as evidenced by reduced blood urea nitrogen (BUN) and serum creatinine (SCr) levels, improved renal histopathology, and decreased expression of renal tubular injury markers such as kidney injury molecule 1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL). Furthermore, olesoxime administration markedly reduced cisplatin-induced apoptosis, inflammation, and oxidative stress in the kidneys of AKI mice. Additionally, olesoxime treatment effectively restored mitochondrial function in the kidneys of AKI mice. In vitro, our results indicated that olesoxime treatment protected against cisplatin-induced apoptosis and mitochondrial dysfunction in cultured HK2 cells. Notably, cisplatin's anticancer effects were unaffected by olesoxime treatment in human cancer cells. CONCLUSION The results of this study suggest that olesoxime is a viable and efficient therapeutic agent in the treatment of cisplatin-induced acute kidney injury presumably by alleviating mitochondrial dysfunction.
Collapse
Affiliation(s)
- Peipei Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Jing Ouyang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Kaiqian Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Yunwen Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Amrein Almira A, Chen MW, El Demerdash N, Javdan C, Park D, Lee JK, Martin LJ. Proteasome localization and activity in pig brain and in vivo small molecule screening for activators. Front Cell Neurosci 2024; 18:1353542. [PMID: 38469354 PMCID: PMC10925635 DOI: 10.3389/fncel.2024.1353542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Loss of proteasome function, proteinopathy, and proteotoxicity may cause neurodegeneration across the human lifespan in several forms of brain injury and disease. Drugs that activate brain proteasomes in vivo could thus have a broad therapeutic impact in neurology. Methods Using pigs, a clinically relevant large animal with a functionally compartmental gyrencephalic cerebral cortex, we evaluated the localization and biochemical activity of brain proteasomes and tested the ability of small molecules to activate brain proteasomes. Results By Western blotting, proteasome protein subunit PSMB5 and PSMA3 levels were similar in different pig brain regions. Immunohistochemistry for PSMB5 showed localization in the cytoplasm (diffuse and particulate) and nucleus (cytoplasm < nucleus). Some PSMB5 immunoreactivity was colocalized with mitochondrial (voltage-gated anion channel and cyclophilin D) and cell death (Aven) proteins in the neuronal soma and neuropil in the neocortex of pig and human brains. In the nucleus, PSMB5 immunoreactivity was diffuse, particulate, and clustered, including perinucleolar decorations. By fluorogenic assay, proteasome chymotrypsin-like activities (CTL) in crude tissue soluble fractions were generally similar within eight different pig brain regions. Proteasome CTL activity in the hippocampus was correlated with activity in nasal mucosa biopsies. In pilot analyses of subcellular fractions of pig cerebral cortex, proteasome CTL activity was highest in the cytosol and then ~50% lower in nuclear fractions; ~15-20% of total CTL activity was in pure mitochondrial fractions. With in-gel activity assay, 26S-singly and -doubly capped proteasomes were the dominant forms in the pig cerebral cortex. With a novel in situ histochemical activity assay, MG132-inhibitable proteasome CTL activity was localized to the neuropil, as a mosaic, and to cell bodies, nuclei, and centrosome-like perinuclear satellites. In piglets treated intravenously with pyrazolone derivative and chlorpromazine over 24 h, brain proteasome CTL activity was modestly increased. Discussion This study shows that the proteasome in the pig brain has relative regional uniformity, prominent nuclear and perinuclear presence with catalytic activity, a mitochondrial association with activity, 26S-single cap dominance, and indications from small molecule systemic administration of pyrazolone derivative and chlorpromazine that brain proteasome function appears safely activable.
Collapse
Affiliation(s)
- Adriana Amrein Almira
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - May W. Chen
- Departments of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nagat El Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cameron Javdan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dongseok Park
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jennifer K. Lee
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lee J. Martin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Sameni M, Moradbeigi P, Hosseini S, Ghaderian SMH, Jajarmi V, Miladipour AH, Basati H, Abbasi M, Salehi M. ZIF-8 Nanoparticle: A Valuable Tool for Improving Gene Delivery in Sperm-Mediated Gene Transfer. Biol Proced Online 2024; 26:4. [PMID: 38279129 PMCID: PMC10811821 DOI: 10.1186/s12575-024-00229-2] [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: 10/21/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024] Open
Abstract
Metal-organic frameworks (MOFs) are porous materials with unique characteristics that make them well-suited for drug delivery and gene therapy applications. Among the MOFs, zeolitic imidazolate framework-8 (ZIF-8) has emerged as a promising candidate for delivering exogenous DNA into cells. However, the potential of ZIF-8 as a vector for sperm-mediated gene transfer (SMGT) has not yet been thoroughly explored.This investigation aimed to explore the potential of ZIF-8 as a vector for enhancing genetic transfer and transgenesis rates by delivering exogenous DNA into sperm cells. To test this hypothesis, we employed ZIF-8 to deliver a plasmid expressing green fluorescent protein (GFP) into mouse sperm cells and evaluated the efficiency of DNA uptake. Our findings demonstrate that ZIF-8 can efficiently load and deliver exogenous DNA into mouse sperm cells, increasing GFP expression in vitro. These results suggest that ZIF-8 is a valuable tool for enhancing genetic transfer in SMGT, with important implications for developing genetically modified animals for research and commercial purposes. Additionally, our study highlights the potential of ZIF-8 as a novel class of vectors for gene delivery in reproductive biology.Overall, our study provides a foundation for further research into using ZIF-8 and other MOFs as gene delivery systems in reproductive biology and underscores the potential of these materials as promising vectors for gene therapy and drug delivery.
Collapse
Affiliation(s)
- Marzieh Sameni
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Moradbeigi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Sara Hosseini
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hasti Noavaran Gene Royan, Tehran, Iran
| | | | - Vahid Jajarmi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Miladipour
- Department of Nephrology, Clinical Research and Development Center at Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojat Basati
- Tissue Engineering Department, TISSUEHUB Co, Tehran, Iran
- Department of Chemical Engineering, Faculty of Engineering, Tehran University, Tehran, Iran
| | - Maryam Abbasi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Zhino-Gene Research Services Co, Tehran, Iran
| | - Mohammad Salehi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Tang H, Yu Y, Zhan X, Chai Y, Zheng Y, Liu Y, Xia D, Lin H. Zeolite imidazolate framework-8 in bone regeneration: A systematic review. J Control Release 2024; 365:558-582. [PMID: 38042375 DOI: 10.1016/j.jconrel.2023.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/19/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
Zeolite imidazolate framework-8 (ZIF-8) is a biomaterial that has been increasingly studied in recent years. It has several applications such as bone regeneration, promotion of angiogenesis, drug loading, and antibacterial activity, and exerts multiple effects to deal with various problems in the process of bone regeneration. This systematic review aims to provide an overview of the applications and effectiveness of ZIF-8 in bone regeneration. A search of papers published in the PubMed, Web of Science, Embase, and Cochrane Library databases revealed 532 relevant studies. Title, abstract, and full-text screening resulted in 39 papers being included in the review, including 39 in vitro and 22 animal studies. Appropriate concentrations of nano ZIF-8 can promote cell proliferation and osteogenic differentiation by releasing Zn2+ and entering the cell, whereas high doses of ZIF-8 are cytotoxic and inhibit osteogenic differentiation. In addition, five studies confirmed that ZIF-8 exhibits good vasogenic activity. In all in vivo experiments, nano ZIF-8 promoted bone formation. These results indicate that, at appropriate concentrations, materials containing ZIF-8 promote bone regeneration more than materials without ZIF-8, and with characteristics such as promoting angiogenesis, drug loading, and antibacterial activity, it is expected to show promising applications in the field of bone regeneration. STATEMENT OF SIGNIFICANCE: This manuscript reviewed the use of ZIF-8 in bone regeneration, clarified the biocompatibility and effectiveness in promoting bone regeneration of ZIF-8 materials, and discussed the possible mechanisms and factors affecting its promotion of bone regeneration. Overall, this study provides a better understanding of the latest advances in the field of bone regeneration of ZIF-8, serves as a design guide, and contributes to the design of future experimental studies.
Collapse
Affiliation(s)
- Hao Tang
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yameng Yu
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Xinxin Zhan
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yuan Chai
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Dandan Xia
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Hong Lin
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| |
Collapse
|
8
|
Tang J, Liu Z, Han J, Xue J, Liu L, Lin J, Wu C, Zhang Q, Wu S, Liu C, Huang H, Fu Y, Li M, Zhuo Y, Li Y. Increased Mobile Zinc Regulates Retinal Ganglion Cell Survival via Activating Mitochondrial OMA1 and Integrated Stress Response. Antioxidants (Basel) 2022; 11:antiox11102001. [PMID: 36290724 PMCID: PMC9598227 DOI: 10.3390/antiox11102001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/26/2022] Open
Abstract
Retinal ganglion cells (RGCs), the projection neurons of the eye, are irreversibly lost once the optic nerve is injured, which is a critical mechanism of glaucoma. Mobile zinc (Zn2+) levels rapidly increase in retinal interneuron amacrine cells and Zn2+ is then transferred to RGCs via the Zn2+ transporter protein ZnT-3, triggering RGC loss in optic nerve injury. Zn2+ chelation and ZnT-3 deletion promote long-term RGC survival. However, the downstream signaling pathways of Zn2+ in RGCs remains unknown. Here, we show that increased levels of Zn2+ upregulate the expression and activity of mitochondrial zinc metallopeptidase OMA1 in the retina, leading to the cleavage of DELE1 and activation of cytosolic eIF2α kinase PKR, triggering the integrated stress response (ISR) in RGCs. Our study identified OMA1 and ISR as the downstream molecular mechanisms of retinal Zn2+ and potential targets for preventing the progression of Zn2+-associated neuronal damage.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhe Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jiaxu Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jingfei Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Liyan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Caiqing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Siting Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Canying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Haishun Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuanyuan Fu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Min Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
- Correspondence: (Y.Z.); (Y.L.)
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
- Correspondence: (Y.Z.); (Y.L.)
| |
Collapse
|
9
|
Huang CH, Wang FT, Chan WH. Role of caspase-3-cleaved/activated PAK2 in brusatol-triggered apoptosis of human lung cancer A549 cells. Toxicol Res (Camb) 2022; 11:791-803. [PMID: 36337251 PMCID: PMC9623572 DOI: 10.1093/toxres/tfac057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 09/01/2023] Open
Abstract
Brusatol, a major quassinoid extract of Bruceae fructus, is an important bioactive component with antineoplastic capacity. Several beneficial pharmacological and biological properties of brusatol have been uncovered to date, including anti-inflammatory, anticolitis, antimalarial, and anticancer activities. To confer anticancer benefits, brusatol is reported to effectively inhibit the Nrf2-mediated antioxidant response and trigger apoptotic signaling. In this study, we investigated the regulatory mechanisms underlying apoptotic processes in brusatol-treated A549 cells in detail. Our experiments showed that brusatol induces cell death through intracellular ROS-triggered mitochondria-dependent apoptotic events and does not involve necrosis. Mechanistically, p21-activated protein kinase 2 (PAK2) was cleaved by caspase-3 to generate an activated p34 fragment involved in brusatol-induced apoptosis of A549 cells. Notably, PAK2 knockdown led to downregulation of caspase-3-mediated PAK2 activity, in turn, effectively attenuating brusatol-induced apoptosis, highlighting a crucial role of caspase-3-activated PAK2 in this process. Moreover, knockdown of PAK2 resulted in significant inhibition of c-Jun N-terminal kinase (JNK) activity in brusatol-treated A549 cells, clearly suggesting that JNK serves as a downstream substrate of caspase-3-cleaved/activated PAK2 in the apoptotic cascade. SP600125, a specific JNK inhibitor, significantly suppressed brusatol-induced JNK activity but only partially prevented apoptosis, implying that JNK serves as only one of a number of substrates for PAK2 in the brusatol-triggered apoptotic cascade. Based on the collective results, we propose a signaling cascade model for brusatol-induced apoptosis in human A549 cells involving ROS, caspases, PAK2, and JNK.
Collapse
Affiliation(s)
- Chien-Hsun Huang
- Department of Obstetrics and Gynecology, Taoyuan General Hospital, Ministry of Health & Welfare, Zhongshan Road, Taoyuan District, Taoyuan City 33004, Taiwan
| | - Fu-Ting Wang
- Rehabilitation and Technical Aid Center, Taipei Veterans General Hospital, Section 2, Shipai Road, Beitou District, Taipei City 11217, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City 32023, Taiwan
| |
Collapse
|
10
|
Pan J, Wei Y, Ni L, Li X, Deng Y, Xu B, Yang T, Sun J, Liu W. Unbalanced ER-mitochondrial calcium homeostasis promotes mitochondrial dysfunction and associated apoptotic pathways activation in methylmercury exposed rat cortical neurons. J Biochem Mol Toxicol 2022; 36:e23136. [PMID: 35678294 DOI: 10.1002/jbt.23136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022]
Abstract
Methylmercury (MeHg) is a cumulative environmental pollutant that can easily cross the blood-brain barrier and cause damage to the brain, mainly targeting the central nervous system. The purpose of this study is to investigate the role of calcium ion (Ca2+ ) homeostasis between the endoplasmic reticulum (ER) and mitochondria in MeHg-induced neurotoxicity. Rat primary cortical neurons exposed to MeHg (0.25-1 μm) underwent dose-dependent cell damage, accompanied by increased Ca2+ release from the ER and elevated levels of free Ca2+ in cytoplasm and mitochondria. MeHg also increased the protein and messenger RNA expressions of the inositol 1,4,5-triphosphate receptor, ryanodine receptor 2, and mitochondrial calcium uniporter. Ca2+ channel inhibitors 2-aminoethyl diphenylborinate and procaine reduced the release of Ca2+ from ER, while RR and 4,4'-diisothiocyanatostilbene-2,2'-disulfonate inhibited Ca2+ uptake from mitochondria. In addition, pretreatment with Ca2+ chelator BAPTA-AM effectively restored mitochondrial membrane potential levels, inhibited over opening of mitochondrial permeability transition pore, and maintained mitochondrial function stability. Meanwhile, the expression of mitochondrial apoptosis-related proteins recovered to some extent, along with the reduction of the early apoptosis ratio. These results suggest that Ca2+ homeostasis plays an essential role in mitochondrial damage and apoptosis induced by MeHg, which may be one of the important mechanisms of MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| | - Jingyi Sun
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
11
|
Liu C, Zeng H, Chen Z, Ge Z, Wang B, Liu B, Fan Z. Sprayable methacrylic anhydride-modified gelatin hydrogel combined with bionic neutrophils nanoparticles for scar-free wound healing of diabetes mellitus. Int J Biol Macromol 2022; 202:418-430. [PMID: 35051497 DOI: 10.1016/j.ijbiomac.2022.01.083] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
Abstract
Hard-to-healing or nonhealing diabetic wounds caused by hyperglycemia, bacterial infection and chronic inflammation are becoming a challenge globally. In this study, a novel hydrogel for diabetic wound healing composed of methacrylic anhydride-modified gelatin (GelMA) hydrogel and mimicking neutrophil nanoparticles was originally created. The prepared GelMA hydrogel has good sprayability and film-formation ability under blue light illumination (wavelength = 435-480 nm). Nanoparticles mimicking neutrophils belong to a double enzyme system that are encapsulated in ZIF-8 nanoparticles, which can consume glucose to produce HClO, ensuring a decrease in the glucose concentration of the wound and growth inhibition in bacteria. The hydrogel also has excellent biocompatibility, which can promote the growth and proliferation of fibroblasts. More importantly, the hydrogel can accelerate wound healing in type I diabetic rats owing to the downregulation of proinflammatory cytokines, and the wound with an area of 1 cm2 can be almost fully healed with no formation of the scar on the 21st day, as verified by histochemistry and immunohistochemistry. All these combinations indicate its potential in diabetic wound treatment.
Collapse
Affiliation(s)
- Changfeng Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Huajing Zeng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Ziyan Chen
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Zhenlin Ge
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Bei Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China.
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
12
|
Song X, Zhang L, Hui X, Sun X, Yang J, Wang J, Wu H, Wang X, Zheng Z, Che F, Wang G. Selenium-containing protein from selenium-enriched Spirulina platensis antagonizes oxygen glucose deprivation-induced neurotoxicity by inhibiting ROS-mediated oxidative damage through regulating MPTP opening. PHARMACEUTICAL BIOLOGY 2021; 59:629-638. [PMID: 34062090 PMCID: PMC8172226 DOI: 10.1080/13880209.2021.1928715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
CONTEXT Selenium-containing protein from selenium-enriched Spirulina platensis (Se-SP) (syn. Arthrospira platensis [Microcoleaceae]) showed novel antioxidant activity. However, the protective effect of Se-SP against oxygen glucose deprivation (OGD)-induced neural apoptosis has not been reported yet. OBJECTIVE To verify whether Se-SP can inhibit OGD-induced neural apoptosis and explore the underlying mechanism. MATERIALS AND METHODS Primary hippocampal neurons were separated from Sprague-Dawley (SD) rats. 95% N2 + 5% CO2 were employed to establish OGD model. Neurons were treated with 5 and 10 µg/mL Se-SP under OGD condition for 6 h. Neurons without treatment were the control group. Neural viability and apoptosis were detected by MTT, immunofluorescence and western blotting methods. RESULTS Se-SP significantly improved neuronal viability (from 57.2% to 94.5%) and inhibited apoptosis in OGD-treated primary neurons (from 45.6% to 6.3%), followed by improved neuronal morphology and caspases activation. Se-SP co-treatment also effectively suppressed OGD-induced DNA damage by inhibiting ROS accumulation in neurons (from 225.6% to 106.3%). Additionally, mitochondrial dysfunction was also markedly improved by Se-SP co-treatment via balancing Bcl-2 family expression. Moreover, inhibition of mitochondrial permeability transition pore (MPTP) by CsA (an MPTP inhibitor) dramatically attenuated OGD-induced ROS generation (from 100% to 56.2%), oxidative damage, mitochondrial membrane potential (MPP) loss (from 7.5% to 44.3%), and eventually reversed the neuronal toxicity and apoptosis (from 57.4% to 79.6%). DISCUSSION AND CONCLUSIONS Se-SP showed enhanced potential to inhibit OGD-induced neurotoxicity and apoptosis by inhibiting ROS-mediated oxidative damage through regulating MPTP opening, indicating that selenium-containing protein showed broad application in the chemoprevention and chemotherapy against human ischaemic brain injury.
Collapse
Affiliation(s)
- Xiaojie Song
- Department of Neurology, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Linyi People’s Hospital, Linyi, China
| | - Lijun Zhang
- Department of Neurology, Linyi People’s Hospital, Linyi, China
| | - Xin Hui
- Department of Neurology, Linyi People’s Hospital, Linyi, China
| | - Xiangfu Sun
- Department of Internal Medicine, Taian Traffic Hospital, Taian, China
| | - Juntao Yang
- Department of Internal Medicine, Taian Traffic Hospital, Taian, China
| | - Jinlei Wang
- Department of Internal Medicine, Taian Traffic Hospital, Taian, China
| | - Hualian Wu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB-CAS), Guangdong Key Laboratory of Marine Materia Medica (LMMM-GD), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Xianjun Wang
- Department of Neurology, Linyi People’s Hospital, Linyi, China
| | - Zuncheng Zheng
- Department of Rehabilitation, Taian City Central Hospital, Taian, ChinaShandong
| | - Fengyuan Che
- Department of Neurology, Linyi People’s Hospital, Linyi, China
| | - Guojun Wang
- Department of Neurosurgery, Taian City Central Hospital, Taian, China
| |
Collapse
|
13
|
Tang J, Zhuo Y, Li Y. Effects of Iron and Zinc on Mitochondria: Potential Mechanisms of Glaucomatous Injury. Front Cell Dev Biol 2021; 9:720288. [PMID: 34447755 PMCID: PMC8383321 DOI: 10.3389/fcell.2021.720288] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Glaucoma is the most substantial cause of irreversible blinding, which is accompanied by progressive retinal ganglion cell damage. Retinal ganglion cells are energy-intensive neurons that connect the brain and retina, and depend on mitochondrial homeostasis to transduce visual information through the brain. As cofactors that regulate many metabolic signals, iron and zinc have attracted increasing attention in studies on neurons and neurodegenerative diseases. Here, we summarize the research connecting iron, zinc, neuronal mitochondria, and glaucomatous injury, with the aim of updating and expanding the current view of how retinal ganglion cells degenerate in glaucoma, which can reveal novel potential targets for neuroprotection.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Wang LL, Du RS, Li J, Cai ZJ, Han L, Mao Y, Zhou YY, Yu QL, Chen LH. The potential mediation of nitric oxide in the activation of mitochondrion-dependent apoptosis and yak meat tenderness during postmortem aging. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
15
|
Martin LJ, Wong M. Skeletal Muscle-Restricted Expression of Human SOD1 in Transgenic Mice Causes a Fatal ALS-Like Syndrome. Front Neurol 2020; 11:592851. [PMID: 33381076 PMCID: PMC7767933 DOI: 10.3389/fneur.2020.592851] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal heterogeneous neurodegenerative disease that causes motor neuron (MN) loss and skeletal muscle paralysis. It is uncertain whether this degeneration of MNs is triggered intrinsically and is autonomous, or if the disease initiating mechanisms are extrinsic to MNs. We hypothesized that skeletal muscle is a primary site of pathogenesis in ALS that triggers MN degeneration. Some inherited forms of ALS are caused by mutations in the superoxide dismutase-1 (SOD1) gene, that encodes an antioxidant protein, so we created transgenic (tg) mice expressing wild-type-, G37R-, and G93A-human SOD1 gene variants only in skeletal muscle. Presence of human SOD1 (hSOD1) protein in skeletal muscle was verified by western blotting, enzyme activity gels, and immunofluorescence in myofibers and satellite cells. These tg mice developed limb weakness and paresis with motor deficits, limb and chest muscle wasting, diaphragm atrophy, and age-related fatal disease with a lifespan shortening of 10–16%. Brown and white adipose tissue also became wasted. Myofibers of tg mice developed crystalline-like inclusions, individualized sarcomere destruction, mitochondriopathy with vesiculation, DNA damage, and activated p53. Satellite cells became apoptotic. The diaphragm developed severe loss of neuromuscular junction presynaptic and postsynaptic integrity, including decreased innervation, loss of synaptophysin, nitration of synaptophysin, and loss of nicotinic acetylcholine receptor and scaffold protein rapsyn. Co-immunoprecipitation identified hSOD1 interaction with rapsyn. Spinal cords of tg mice developed gross atrophy. Spinal MNs formed cytoplasmic and nuclear inclusions, axonopathy, mitochondriopathy, accumulated DNA damage, activated p53 and cleaved caspase-3, and died. Tg mice had a 40–50% loss of MNs. This work shows that hSOD1 in skeletal muscle is a driver of pathogenesis in ALS, that involves myofiber and satellite cell toxicity, and apparent muscle-adipose tissue disease relationships. It also identifies a non-autonomous mechanism for MN degeneration explaining their selective vulnerability as likely a form of target-deprivation retrograde neurodegeneration.
Collapse
Affiliation(s)
- Lee J Martin
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Margaret Wong
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Zakyrjanova GF, Gilmutdinov AI, Tsentsevitsky AN, Petrov AM. Olesoxime, a cholesterol-like neuroprotectant restrains synaptic vesicle exocytosis in the mice motor nerve terminals: Possible role of VDACs. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158739. [PMID: 32428575 DOI: 10.1016/j.bbalip.2020.158739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
Olesoxime is a cholesterol-like neuroprotective compound that targets to mitochondrial voltage dependent anion channels (VDACs). VDACs were also found in the plasma membrane and highly expressed in the presynaptic compartment. Here, we studied the effects of olesoxime and VDAC inhibitors on neurotransmission in the mouse neuromuscular junction. Electrophysiological analysis revealed that olesoxime suppressed selectively evoked neurotransmitter release in response to a single stimulus and 20 Hz activity. Also olesoxime decreased the rate of FM1-43 dye loss (an indicator of synaptic vesicle exocytosis) at low frequency stimulation and 20 Hz. Furthermore, an increase in extracellular Cl- enhanced the action of olesoxime on the exocytosis and olesoxime increased intracellular Cl- levels. The effects of olesoxime on the evoked synaptic vesicle exocytosis and [Cl-]i were blocked by membrane-permeable and impermeable VDAC inhibitors. Immunofluorescent labeling pointed on the presence of VDACs on the synaptic membranes. Rotenone-induced mitochondrial dysfunction perturbed the exocytotic release of FM1-43 and cell-permeable VDAC inhibitor (but not olesoxime or impermeable VDAC inhibitor) partially mitigated the rotenone-driven alterations in the FM1-43 unloading and mitochondrial superoxide production. Thus, olesoxime restrains neurotransmission by acting on plasmalemmal VDACs whose activation can limit synaptic vesicle exocytosis probably via increasing anion flux into the nerve terminals.
Collapse
Affiliation(s)
- Guzalia F Zakyrjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan 420012, Russia
| | - Amir I Gilmutdinov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia
| | - Andrey N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan 420012, Russia.
| |
Collapse
|
17
|
Eckert GP, Eckert SH, Eckmann J, Hagl S, Muller WE, Friedland K. Olesoxime improves cerebral mitochondrial dysfunction and enhances Aβ levels in preclinical models of Alzheimer's disease. Exp Neurol 2020; 329:113286. [PMID: 32199815 DOI: 10.1016/j.expneurol.2020.113286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/15/2020] [Accepted: 03/17/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Approved drugs for Alzheimer's disease (AD) only have a symptomatic effects and do not intervene causally in the course of the disease. Olesoxime (TRO19622) has been tested in AD disease models characterized by improved amyloid precursor protein processing (AβPP) and mitochondrial dysfunction. METHODS Three months old Thy-1-AβPPSL (tg) and wild type mice (wt) received TRO19622 (100 mg/kg b.w.) in supplemented food pellets for 15 weeks (tg TRO19622). Mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP) levels were determined in dissociated brain cells (DBC). Respiration was analyzed in mitochondria isolated from brain tissue. Citrate synthase (CS) activity and beta-amyloid peptide (Aβ1-40) levels were determined in brain tissue. Malondialdehyde (MDA) levels were determined as an indicator for lipid peroxidation. DBC and brain homogenates were additionally stressed with Rotenone and FeCl2, respectively. Mitochondrial respiration and Aβ1-40 levels were also determined in HEK-AβPPsw-cells. RESULTS Treatment of mice did not affect the body weight. TRO19622 was absorbed after oral treatment (plasma levels: 6,2 μg/ml). Mitochondrial respiration was significantly reduced in brains of tg-mice. Subsequently, DBC isolated from brains of tg-mice showed significantly lower MMP but not ATP levels. TRO19622 increased the activity of respiratory chain complexes and reversed complex IV (CIV) activity and MMP. Moreover, DBC isolated from brains of tg TRO19622 mice were protected from Rotenone induced inhibition of complex I activity. TRO19622 also increased the respiratory activity in HEKsw-cells. MDA basal levels were significantly higher in brain homogenates isolated from tg-mice. TRO19622 treatment had no effects on lipid peroxidation. TRO19622 increased cholesterol levels but did not change membrane fluidity of synaptosomal plasma and mitochondrial membranes isolated from brain of mice. TRO19622 significantly increased levels of Aβ1-40 in both, in brains of tg TRO19622 mice and in HEKsw cells. CONCLUSIONS TRO19622 improves mitochondrial dysfunction but enhances Aβ levels in disease models of AD. Further studies must evaluate whether TRO19622 offers benefits at the mitochondrial level despite the increased formation of Aβ, which could be harmful.
Collapse
Affiliation(s)
- Gunter P Eckert
- Institute of Nutritional Sciences, Justus-Liebig-University, Giessen, Germany.
| | - Schamim H Eckert
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Janett Eckmann
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Stephanie Hagl
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Walter E Muller
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Kristina Friedland
- Institute of Pharmacology, Johannes-Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Kim BW, Jeong YE, Wong M, Martin LJ. DNA damage accumulates and responses are engaged in human ALS brain and spinal motor neurons and DNA repair is activatable in iPSC-derived motor neurons with SOD1 mutations. Acta Neuropathol Commun 2020; 8:7. [PMID: 32005289 PMCID: PMC6995159 DOI: 10.1186/s40478-019-0874-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
DNA damage is implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). However, relationships between DNA damage accumulation, DNA damage response (DDR), and upper and lower motor neuron vulnerability in human ALS are unclear; furthermore, it is unknown whether epigenetic silencing of DNA repair pathways contributes to ALS pathogenesis. We tested the hypotheses that DNA damage accumulates in ALS motor neurons along with diminished DDR, and that DNA repair genes undergo hypermethylation. Human postmortem CNS tissue was obtained from ALS cases (N = 34) and age-matched controls without neurologic disease (N = 15). Compared to age-matched controls, abasic sites accumulated in genomic DNA of ALS motor cortex and laser capture microdissection-acquired spinal motor neurons but not in motor neuron mitochondrial DNA. By immunohistochemistry, DNA damage accumulated significantly in upper and lower motor neurons in ALS cases as single-stranded DNA and 8-hydroxy-deoxyguanosine (OHdG) compared to age-matched controls. Significant DDR was engaged in ALS motor neurons as evidenced by accumulation of c-Abl, nuclear BRCA1, and ATM activation. DNA damage and DDR were present in motor neurons at pre-attritional stages and throughout the somatodendritic attritional stages of neurodegeneration. Motor neurons with DNA damage were also positive for activated p53 and cleaved caspase-3. Gene-specific promoter DNA methylation pyrosequencing identified the DNA repair genes Ogg1, Apex1, Pnkp and Aptx as hypomethylated in ALS. In human induced-pluripotent stem cell (iPSC)-derived motor neurons with familial ALS SOD1 mutations, DNA repair capacity was similar to isogenic control motor neurons. Our results show that vulnerable neurons in human ALS accumulate DNA damage, and contrary to our hypothesis, strongly activate and mobilize response effectors and DNA repair genes. This DDR in ALS motor neurons involves recruitment of c-Abl and BRCA1 to the nucleus in vivo, and repair of DNA double-strand breaks in human ALS motor neurons with SOD1 mutations in cell culture.
Collapse
Affiliation(s)
- Byung Woo Kim
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ye Eun Jeong
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA
| | - Lee J Martin
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA.
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Vasquez-Vivar J, Shi Z, Jeong JW, Luo K, Sharma A, Thirugnanam K, Tan S. Neuronal vulnerability to fetal hypoxia-reoxygenation injury and motor deficit development relies on regional brain tetrahydrobiopterin levels. Redox Biol 2020; 29:101407. [PMID: 31926630 PMCID: PMC6928344 DOI: 10.1016/j.redox.2019.101407] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/13/2019] [Accepted: 12/09/2019] [Indexed: 11/29/2022] Open
Abstract
Hypertonia is pathognomonic of cerebral palsy (CP), often caused by brain injury before birth. To understand the early driving events of hypertonia, we utilized magnetic resonance imaging (MRI) assessment of early critical brain injury in rabbit fetuses (79% term) that will predict hypertonia after birth following antenatal hypoxia-ischemia. We examined if individual variations in the tetrahydrobiopterin cofactor in the parts of the brain controlling motor function could indicate a role in specific damage to motor regions and disruption of circuit integration as an underlying mechanism for acquiring motor disorders, which has not been considered before. The rabbit model mimicked acute placental insufficiency and used uterine ischemia at a premature gestation. MRI during the time of hypoxia-ischemia was used to differentiate which individual fetal brains would become hypertonic. Four brain regions collected immediately after hypoxia-ischemia or 48 h later were analyzed in a blinded fashion. Age-matched sham-operated animals were used as controls. Changes in the reactive nitrogen species and gene expression of the tetrahydrobiopterin biosynthetic enzymes in brain regions were also studied. We found that a combination of low tetrahydrobiopterin content in the cortex, basal ganglia, cerebellum, and thalamus brain regions, but not a unique low threshold of tetrahydrobiopterin, contributed etiologically to hypertonia. The biggest contribution was from the thalamus. Evidence for increased reactive nitrogen species was found in the cortex. By 48 h, tetrahydrobiopterin and gene expression levels in the different parts of the brain were not different between MRI stratified hypertonia and non-hypertonia groups. Sepiapterin treatment given to pregnant dams immediately after hypoxia-ischemia ameliorated hypertonia and death. We conclude that a developmental tetrahydrobiopterin variation is necessary with fetal hypoxia-ischemia and is critical for disrupting normal motor circuits that develop into hypertonia. The possible mechanistic pathway involves reactive nitrogen species.
Collapse
Affiliation(s)
- Jeannette Vasquez-Vivar
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhongjie Shi
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeong-Won Jeong
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA; Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kehuan Luo
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Amit Sharma
- Neonatology Division, Children's Hospital of Michigan, Detroit, MI, USA
| | - Karthikeyan Thirugnanam
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sidhartha Tan
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA; Neonatology Division, Children's Hospital of Michigan, Detroit, MI, USA.
| |
Collapse
|
20
|
Weber JJ, Clemensson LE, Schiöth HB, Nguyen HP. Olesoxime in neurodegenerative diseases: Scrutinising a promising drug candidate. Biochem Pharmacol 2019; 168:305-318. [PMID: 31283931 DOI: 10.1016/j.bcp.2019.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022]
Abstract
Over the last years, the experimental compound olesoxime, a mitochondria-targeting cholesterol derivative, has emerged as a promising drug candidate for neurodegenerative diseases. Numerous preclinical studies have successfully proved olesoxime's neuroprotective properties in cell and animal models of clinical conditions such as amyotrophic lateral sclerosis, Huntington disease, Parkinson disease, peripheral neuropathy and spinal muscular atrophy. The beneficial effects were attributed to olesoxime's potential impact on oxidative stress, mitochondrial permeability transition or cholesterol homoeostasis. Although no significant benefits have been demonstrated in patients of amyotrophic lateral sclerosis, and only the first 12 months of a phase II/III clinical trial showed an improvement in motor symptoms of spinal muscular atrophy, this orphan drug may still offer undiscovered potential in the treatment of neurological diseases. In our earlier preclinical studies, we demonstrated that administration of olesoxime in mouse and rat models of Huntington disease improved psychiatric and molecular phenotypes. Aside from stabilising mitochondrial function, the drug reduced the overactivation of calpains, a class of calcium-dependent proteases entangled in neurodegenerative conditions. This observation may be credited to olesoxime's action on calcium dyshomeostasis, a further hallmark in neurodegeneration, and linked to its targets TSPO and VDAC, two proteins of the outer mitochondrial membrane associated with mitochondrial calcium handling. Further research into the mode of action of olesoxime under pathological conditions, including its effect on neuronal calcium homeostasis, may strengthen the untapped potential of olesoxime or other similar compounds as a therapeutic for neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonasz Jeremiasz Weber
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.
| | | | - Helgi Birgir Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden; Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
21
|
Muckova L, Vanova N, Misik J, Herman D, Pejchal J, Jun D. Oxidative stress induced by oxime reactivators of acetylcholinesterase in vitro. Toxicol In Vitro 2019; 56:110-117. [DOI: 10.1016/j.tiv.2019.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 10/27/2022]
|
22
|
Bisicchia E, Sasso V, Molinari M, Viscomi MT. Plasticity of microglia in remote regions after focal brain injury. Semin Cell Dev Biol 2019; 94:104-111. [PMID: 30703556 DOI: 10.1016/j.semcdb.2019.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
The CNS is endowed with an intrinsic ability to recover from and adapt secondary compensatory mechanisms to injury. The basis of recovery stems from brain plasticity, defined as the brain's ability to make adaptive changes on structural and functional levels, ranging from molecular, synaptic, and cellular changes in response to alterations in their environment. In this multitude of responses, microglia have an active role and contribute to brain plasticity through their dynamic responses. This review will provide an overview of microglial responses in the context of acute CNS injury and their function in post-traumatic repair and assess the changes that are induced by damage in remote areas from, but functionally connected to, the primary site of injury. In the second section, we highlight the effects of several therapeutic approaches, with particular interest paid to specialized pro-resolving lipid mediators, in modulating microglial responses in remote regions and enhancing long-term functional recovery via suppression of neurodegenerative cascades that are induced by damage, which may contribute to a translational bridge from bench to bedside.
Collapse
Affiliation(s)
- Elisa Bisicchia
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valeria Sasso
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marco Molinari
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Teresa Viscomi
- Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del S. Cuore, Rome, Italy.
| |
Collapse
|
23
|
Yang M, Xu Y, Heisner JS, Sun J, Stowe DF, Kwok WM, Camara AKS. Peroxynitrite nitrates adenine nucleotide translocase and voltage-dependent anion channel 1 and alters their interactions and association with hexokinase II in mitochondria. Mitochondrion 2018; 46:380-392. [PMID: 30391711 DOI: 10.1016/j.mito.2018.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/26/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
Cardiac ischemia and reperfusion (IR) injury induces excessive emission of deleterious reactive O2 and N2 species (ROS/RNS), including the non-radical oxidant peroxynitrite (ONOO-) that can cause mitochondria dysfunction and cell death. In this study, we explored whether IR injury in isolated hearts induces tyrosine nitration of adenine nucleotide translocase (ANT) and alters its interaction with the voltage-dependent anion channel 1 (VDAC1). We found that IR injury induced tyrosine nitration of ANT and that exposure of isolated cardiac mitochondria to ONOO- induced ANT tyrosine, Y81, nitration. The exposure of isolated cardiac mitochondria to ONOO- also led ANT to form high molecular weight proteins and dissociation of ANT from VDAC1. We found that IR injury in isolated hearts, hypoxic injury in H9c2 cells, and ONOO- treatment of H9c2 cells and isolated mitochondria, each decreased mitochondrial bound-hexokinase II (HK II), which suggests that ONOO- caused HK II to dissociate from mitochondria. Moreover, we found that mitochondria exposed to ONOO- induced VDAC1 oligomerization which may decrease its binding with HK II. We have reported that ONOO- produced during cardiac IR injury induced tyrosine nitration of VDAC1, which resulted in conformational changes of the protein and increased channel conductance associated with compromised cardiac function on reperfusion. Thus, our results imply that ONOO- produced during IR injury and hypoxic stress impeded HK II association with VDAC1. ONOO- exposure nitrated mitochondrial proteins and also led to cytochrome c (cyt c) release from mitochondria. In addition, in isolated mitochondria exposed to ONOO- or obtained after IR, there was significant compromise in mitochondrial respiration and delayed repolarization of membrane potential during oxidative (ADP) phosphorylation. Taken together, ONOO- produced during cardiac IR injury can nitrate tyrosine residues of two key mitochondrial membrane proteins involved in bioenergetics and energy transfer to contribute to mitochondrial and cellular dysfunction.
Collapse
Affiliation(s)
- Meiying Yang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yanji Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China
| | - James S Heisner
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jie Sun
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Institute of Clinical Medicine Research, Suzhou Hospital affiliated with Nanjing Medical University, Suzhou, Jiangsu, China; Department of Gastroenterology and Hepatology, Suzhou Hospital affiliated with Nanjing Medical University, Suzhou, Jiangsu, China
| | - David F Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Research Service, Zablocki VA Medical Center, Milwaukee, WI, USA
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
24
|
Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC. Neuronal Cell Death. Physiol Rev 2018; 98:813-880. [PMID: 29488822 PMCID: PMC5966715 DOI: 10.1152/physrev.00011.2017] [Citation(s) in RCA: 726] [Impact Index Per Article: 103.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Neuronal cell death occurs extensively during development and pathology, where it is especially important because of the limited capacity of adult neurons to proliferate or be replaced. The concept of cell death used to be simple as there were just two or three types, so we just had to work out which type was involved in our particular pathology and then block it. However, we now know that there are at least a dozen ways for neurons to die, that blocking a particular mechanism of cell death may not prevent the cell from dying, and that non-neuronal cells also contribute to neuronal death. We review here the mechanisms of neuronal death by intrinsic and extrinsic apoptosis, oncosis, necroptosis, parthanatos, ferroptosis, sarmoptosis, autophagic cell death, autosis, autolysis, paraptosis, pyroptosis, phagoptosis, and mitochondrial permeability transition. We next explore the mechanisms of neuronal death during development, and those induced by axotomy, aberrant cell-cycle reentry, glutamate (excitoxicity and oxytosis), loss of connected neurons, aggregated proteins and the unfolded protein response, oxidants, inflammation, and microglia. We then reassess which forms of cell death occur in stroke and Alzheimer's disease, two of the most important pathologies involving neuronal cell death. We also discuss why it has been so difficult to pinpoint the type of neuronal death involved, if and why the mechanism of neuronal death matters, the molecular overlap and interplay between death subroutines, and the therapeutic implications of these multiple overlapping forms of neuronal death.
Collapse
Affiliation(s)
- Michael Fricker
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Aviva M Tolkovsky
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Vilmante Borutaite
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Michael Coleman
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Guy C Brown
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
25
|
Purroy R, Britti E, Delaspre F, Tamarit J, Ros J. Mitochondrial pore opening and loss of Ca 2+ exchanger NCLX levels occur after frataxin depletion. Biochim Biophys Acta Mol Basis Dis 2018; 1864:618-631. [PMID: 29223733 DOI: 10.1016/j.bbadis.2017.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/29/2017] [Accepted: 12/05/2017] [Indexed: 12/12/2022]
Abstract
Frataxin-deficient neonatal rat cardiomyocytes and dorsal root ganglia neurons have been used as cell models of Friedreich ataxia. In previous work we show that frataxin depletion resulted in mitochondrial swelling and lipid droplet accumulation in cardiomyocytes, and compromised DRG neurons survival. Now, we show that these cells display reduced levels of the mitochondrial calcium transporter NCLX that can be restored by calcium-chelating agents and by external addition of frataxin fused to TAT peptide. Also, the transcription factor NFAT3, involved in cardiac hypertrophy and apoptosis, becomes activated by dephosphorylation in both cardiomyocytes and DRG neurons. In cardiomyocytes, frataxin depletion also results in mitochondrial permeability transition pore opening. Since the pore opening can be inhibited by cyclosporin A, we show that this treatment reduces lipid droplets and mitochondrial swelling in cardiomyocytes, restores DRG neuron survival and inhibits NFAT dephosphorylation. These results highlight the importance of calcium homeostasis and that targeting mitochondrial pore by repurposing cyclosporin A, could be envisaged as a new strategy to treat the disease.
Collapse
Affiliation(s)
- R Purroy
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - E Britti
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - F Delaspre
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - J Tamarit
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - J Ros
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain.
| |
Collapse
|
26
|
Javadov S, Jang S, Parodi-Rullán R, Khuchua Z, Kuznetsov AV. Mitochondrial permeability transition in cardiac ischemia-reperfusion: whether cyclophilin D is a viable target for cardioprotection? Cell Mol Life Sci 2017; 74:2795-2813. [PMID: 28378042 PMCID: PMC5977999 DOI: 10.1007/s00018-017-2502-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 12/13/2022]
Abstract
Growing number of studies provide strong evidence that the mitochondrial permeability transition pore (PTP), a non-selective channel in the inner mitochondrial membrane, is involved in the pathogenesis of cardiac ischemia-reperfusion and can be targeted to attenuate reperfusion-induced damage to the myocardium. The molecular identity of the PTP remains unknown and cyclophilin D is the only protein commonly accepted as a major regulator of the PTP opening. Therefore, cyclophilin D is an attractive target for pharmacological or genetic therapies to reduce ischemia-reperfusion injury in various animal models and humans. Most animal studies demonstrated cardioprotective effects of PTP inhibition; however, a recent large clinical trial conducted by international groups demonstrated that cyclosporine A, a cyclophilin D inhibitor, failed to protect the heart in patients with myocardial infarction. These studies, among others, raise the question of whether cyclophilin D, which plays an important physiological role in the regulation of cell metabolism and mitochondrial bioenergetics, is a viable target for cardioprotection. This review discusses previous studies to provide comprehensive information on the physiological role of cyclophilin D as well as PTP opening in the cell that can be taken into consideration for the development of new PTP inhibitors.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Rebecca Parodi-Rullán
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Zaza Khuchua
- Cincinnati Children's Research Foundation, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH, 54229, USA
| | - Andrey V Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
27
|
Salameh A, Keller M, Dähnert I, Dhein S. Olesoxime Inhibits Cardioplegia-Induced Ischemia/Reperfusion Injury. A Study in Langendorff-Perfused Rabbit Hearts. Front Physiol 2017; 8:324. [PMID: 28579963 PMCID: PMC5437207 DOI: 10.3389/fphys.2017.00324] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/05/2017] [Indexed: 12/15/2022] Open
Abstract
Objective: During cardioplegia, which is often used in cardiac surgery, the heart is subjected to global ischemia/reperfusion injury, which can result in a post-operative impairment of cardiac function. Mitochondria permeability transition pores (MPTP) play a key role in cardiomyocyte survival after ischemia/reperfusion injury. It was shown in clinical settings that blockers of MPTP like cyclosporine might have a positive influence on cardiac function after cardioplegic arrest. Olesoxime, which is a new drug with MPTP blocking activity, has been introduced as a neuroprotective therapeutic agent. This drug has not been investigated on a possible positive effect in ischemia/reperfusion injury in hearts. Therefore, the aim of our study was to investigate possible effects of olesoxime on cardiac recovery after cardioplegic arrest. Methods: We evaluated 14 mature Chinchilla bastard rabbits of 1,500–2,000 g. Rabbit hearts were isolated and perfused with constant pressure according to Langendorff. After induction of cardioplegic arrest (30 ml 4°C cold Custodiol cardioplegia without and with 5 μmol/L olesoxime, n = 7 each) the hearts maintained arrested for 90-min. Thereafter, the hearts were re-perfused for 60 min. At the end of each experiment left ventricular samples were frozen in liquid nitrogen for ATP measurements. Furthermore, heart slices were embedded in paraffin for histological analysis. During the entire experiment hemodynamic and functional data such as left ventricular pressure (LVP), dp/dt(max) and (min), pressure rate product (PRP), coronary flow, pO2, and pCO2 were also assessed. Results: Histological analysis revealed that despite the same ischemic burden for both groups markers of nitrosative and oxidative stress were significantly lower in the olesoxime group. Moreover, hearts of the olesoxime-group showed a significantly faster and better hemodynamic recovery during reperfusion. In addition, tissue ATP-levels were significantly higher in the olesoxime treated hearts. Conclusions: Olesoxime significantly protected the cardiac muscle from ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Aida Salameh
- Clinic for Pediatric Cardiology, Heart Centre, University of LeipzigLeipzig, Germany
| | - Maren Keller
- Clinic for Pediatric Cardiology, Heart Centre, University of LeipzigLeipzig, Germany
| | - Ingo Dähnert
- Clinic for Pediatric Cardiology, Heart Centre, University of LeipzigLeipzig, Germany
| | - Stefan Dhein
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of LeipzigLeipzig, Germany
| |
Collapse
|
28
|
Venegoni W, Shen Q, Thimmesch AR, Bell M, Hiebert JB, Pierce JD. The use of antioxidants in the treatment of traumatic brain injury. J Adv Nurs 2017; 73:1331-1338. [PMID: 28103389 DOI: 10.1111/jan.13259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 11/26/2022]
Abstract
AIMS The aim of this study was to discuss secondary traumatic brain injury, the mitochondria and the use of antioxidants as a treatment. BACKGROUND One of the leading causes of death globally is traumatic brain injury, affecting individuals in all demographics. Traumatic brain injury is produced by an external blunt force or penetration resulting in alterations in brain function or pathology. Often, with a traumatic brain injury, secondary injury causes additional damage to the brain tissue that can have further impact on recovery and the quality of life. Secondary injury occurs when metabolic and physiologic processes alter after initial injury and includes increased release of toxic free radicals that cause damage to adjacent tissues and can eventually lead to neuronal necrosis. Although antioxidants in the tissues can reduce free radical damage, the magnitude of increased free radicals overwhelms the body's reduced defence mechanisms. Supplementing the body's natural supply of antioxidants, such as coenzyme Q10, can attenuate oxidative damage caused by reactive oxygen species. DESIGN Discussion paper. DATA SOURCES Research literature published from 2011-2016 in PubMed, CINAHL and Cochrane. IMPLICATIONS FOR NURSING Prompt and accurate assessment of patients with traumatic brain injury by nurses is important to ensure optimal recovery and reduced lasting disability. Thus, it is imperative that nurses be knowledgeable about the secondary injury that occurs after a traumatic brain injury and aware of possible antioxidant treatments. CONCLUSION The use of antioxidants has potential to reduce the magnitude of secondary injury in patients who experience a traumatic brain injury.
Collapse
Affiliation(s)
| | - Qiuhua Shen
- School of Nursing, University of Kansas, Kansas, USA
| | | | - Meredith Bell
- School of Nursing, University of Kansas, Kansas, USA
| | | | | |
Collapse
|
29
|
Magalon K, Le Grand M, El Waly B, Moulis M, Pruss R, Bordet T, Cayre M, Belenguer P, Carré M, Durbec P. Olesoxime favors oligodendrocyte differentiation through a functional interplay between mitochondria and microtubules. Neuropharmacology 2016; 111:293-303. [DOI: 10.1016/j.neuropharm.2016.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/18/2016] [Accepted: 09/07/2016] [Indexed: 11/25/2022]
|
30
|
Martin LJ, Wong M. Enforced DNA repair enzymes rescue neurons from apoptosis induced by target deprivation and axotomy in mouse models of neurodegeneration. Mech Ageing Dev 2016; 161:149-162. [PMID: 27364693 DOI: 10.1016/j.mad.2016.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/23/2016] [Accepted: 06/26/2016] [Indexed: 02/06/2023]
Abstract
It is unknown whether DNA damage accumulation is an upstream instigator or secondary effect of the cell death process in different populations of adult postmitotic neurons in the central nervous system. In two different mouse models of injury-induced neurodegeneration characterized by relatively synchronous accumulation of mitochondria, oxidative stress, and DNA damage prior to neuronal apoptosis, we enforced the expression of human 8-oxoguanine DNA glycosylase (hOGG1) and human apurinic-apyrimidinic endonuclease-1/Ref1 (hAPE) using recombinant adenoviruses (Ad). Thalamic lateral geniculate neurons and lumbar spinal cord motor neurons were transduced by Ad-hOGG1 and Ad-hAPE injections into the occipital cortex and skeletal muscle, respectively, prior to their target deprivation- and axotomy-induced retrograde apoptosis. Enforced expression of hOGG1 and hAPE in thalamus and spinal cord was confirmed by western blotting and immunohistochemistry. In injured populations of neurons in thalamus and spinal cord, a DNA damage response (DDR) was registered, as shown by localization of phospho-activated p53, Rad17, and replication protein A-32 immunoreactivities, and this DDR was attenuated more effectively by enforced hAPE expression than by hOGG1 expression. Enforced expression of hOGG1 and hAPE significantly protected thalamic neurons and motor neurons from retrograde apoptosis induced by target deprivation and axotomy. We conclude that a DDR response is engaged pre-apoptotically in different types of injured mature CNS neurons and that DNA repair enzymes can regulate the survival of retrogradely dying neurons, suggesting that DNA damage and activation of DDR are upstream mechanisms for this form of adult neurodegeneration in vivo, thus identifying DNA repair as a therapeutic target for neuroprotection.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Margaret Wong
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
31
|
Ogundele OM, Wasiu Gbolahan B, Emmanuel Cobham A, Azeez Olakunle I, Abdulbasit A. Differential oxidative stress thresholds distinguishes cellular response to vascular occlusion and chemotoxicityin vivo. Drug Chem Toxicol 2016; 40:101-109. [DOI: 10.1080/01480545.2016.1188300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Kasimov MR, Zakyrjanova GF, Giniatullin AR, Zefirov AL, Petrov AM. Similar oxysterols may lead to opposite effects on synaptic transmission: Olesoxime versus 5α-cholestan-3-one at the frog neuromuscular junction. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:606-16. [PMID: 27102612 DOI: 10.1016/j.bbalip.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/17/2016] [Accepted: 04/15/2016] [Indexed: 02/03/2023]
Abstract
Cholesterol oxidation products frequently have a high biological activity. In the present study, we have used microelectrode recording of end plate currents and FM-based optical detection of synaptic vesicle exo-endocytosis to investigate the effects of two structurally similar oxysterols, olesoxime (cholest-4-en-3-one, oxime) and 5ɑ-cholestan-3-one (5ɑCh3), on neurotransmission at the frog neuromuscular junction. Olesoxime is an exogenous, potentially neuroprotective, substance and 5ɑCh3 is an intermediate product in cholesterol metabolism, which is elevated in the case of cerebrotendinous xanthomatosis. We found that olesoxime slightly increased evoked neurotransmitter release in response to a single stimulus and significantly reduced synaptic depression during high frequency activity. The last effect was due to an increase in both the number of synaptic vesicles involved in exo-endocytosis and the rate of synaptic vesicle recycling. In contrast, 5ɑCh3 reduced evoked neurotransmitter release during the low- and high frequency synaptic activities. The depressant action of 5ɑCh3 was associated with a reduction in the number of synaptic vesicles participating in exo- and endocytosis during high frequency stimulation, without a change in rate of the synaptic vesicle recycling. Of note, olesoxime increased the staining of synaptic membranes with the B-subunit of cholera toxin and the formation of fluorescent ganglioside GM1 clusters, and decreased the fluorescence of 22-NBD-cholesterol, while 5ɑCh3 had the opposite effects, suggesting that the two oxysterols have different effects on lipid raft stability. Taken together, these data show that these two structurally similar oxysterols induce marked different changes in neuromuscular transmission which are related with the alteration in synaptic vesicle cycle.
Collapse
Affiliation(s)
- M R Kasimov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - G F Zakyrjanova
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A R Giniatullin
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A L Zefirov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A M Petrov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia.
| |
Collapse
|
33
|
Schwann Cell and Axon: An Interlaced Unit—From Action Potential to Phenotype Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:183-201. [DOI: 10.1007/978-3-319-40764-7_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Graham RM, Thompson JW, Webster KA. BNIP3 promotes calcium and calpain-dependent cell death. Life Sci 2015; 142:26-35. [PMID: 26471219 DOI: 10.1016/j.lfs.2015.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/24/2015] [Accepted: 10/09/2015] [Indexed: 11/15/2022]
Abstract
AIMS Loss of cardiac muscle by programmed cell death contributes to the progression of ischemic heart disease. Hypoxia, metabolite waste buildup and energy depletion are components of ischemia which may initiate caspase dependent and independent cell death pathways. Previous work from our laboratory has shown that combined hypoxia with acidosis, a hallmark of ischemia promotes cardiac myocyte injury with increasing severity as the pH declines. Hypoxia-acidosis was demonstrated to activate the pro-apoptotic Bcl-2 protein BNIP3 which initiated opening of the mitochondrial permeability transition pore and cell death in the absence of caspase activation. Because calpains are known to contribute to ischemic myocardial damage in some models, we hypothesized that they are intermediates in the BNIP3-mediated death caused by hypoxia-acidosis. MAIN METHODS Neonatal rat cardiac myocytes were subjected to hypoxia with and without acidosis and the contribution of calpains to hypoxia-acidosis cell death determined. KEY FINDINGS Here we report that the death pathway activated by hypoxia-acidosis is driven by a combination of calcium-activated calpains and pro-death factors (DNases) secreted by the mitochondria. Cytochrome c accumulated in the cytoplasm during hypoxia-acidosis but caspase activity was repressed through a calpain-dependent process that prevents the cleavage of procaspase 3. Calpain inhibitors provide vigorous protection against hypoxia-acidosis-induced programmed death. Knockdown of BNIP3 with siRNA prevented calpain activation confirming a central role of BNIP3 in this pathway. SIGNIFICANCE The results implicate BNIP3 and calpain as dependent components of cardiac myocyte death caused by hypoxia-acidosis.
Collapse
Affiliation(s)
- Regina M Graham
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - John W Thompson
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology, Vascular Biology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
35
|
Bnip3 Binds and Activates p300: Possible Role in Cardiac Transcription and Myocyte Morphology. PLoS One 2015; 10:e0136847. [PMID: 26317696 PMCID: PMC4552727 DOI: 10.1371/journal.pone.0136847] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/17/2015] [Indexed: 12/04/2022] Open
Abstract
Bnip3 is a hypoxia-regulated member of the Bcl-2 family of proteins that is implicated in apoptosis, programmed necrosis, autophagy and mitophagy. Mitochondria are thought to be the primary targets of Bnip3 although its activities may extend to the ER, cytoplasm, and nucleus. Bnip3 is induced in the heart by ischemia and pressure-overload, and may contribute to cardiomyopathy and heart failure. Only mitochondrial-dependent programmed death actions have been described for Bnip3 in the heart. Here we describe a novel activity of Bnip3 in cultured cardiac myocytes and transgenic mice overexpressing Bnip3 in the heart (Bnip3-TG). In cultured myocytes Bnip3 bound and activated the acetyltransferase p300, increased acetylation of histones and the transcription factor GATA4, and conferred p300 and GATA4-sensitive cellular morphological changes. In intact Bnip3-TG hearts Bnip3 also bound p300 and GATA4 and conferred enhanced GATA4 acetylation. Bnip3-TG mice underwent age-dependent ventricular dilation and heart failure that was partially prevented by p300 inhibition with curcumin. The results suggest that Bnip3 regulates cardiac gene expression and perhaps myocyte morphology by activating nuclear p300 acetyltransferase activity and hyperacetylating histones and p300-selective transcription factors.
Collapse
|
36
|
Gouarné C, Giraudon-Paoli M, Seimandi M, Biscarrat C, Tardif G, Pruss RM, Bordet T. Olesoxime protects embryonic cortical neurons from camptothecin intoxication by a mechanism distinct from BDNF. Br J Pharmacol 2015; 168:1975-88. [PMID: 23278424 DOI: 10.1111/bph.12094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Olesoxime is a small cholesterol-oxime promoting rat embryonic motor neurons survival in the absence of trophic factors. Because olesoxime can substitute for neurotrophic factors in many situations, and to gain further understanding of its mechanism of action, we wondered if it could prevent neuronal death induced by camptothecin (CPT) and compared its effects with those of brain-derived neurotrophic factor (BDNF). EXPERIMENTAL APPROACH E17 rat embryonic cortical neurons were treated with olesoxime, BDNF or vehicle and intoxicated with CPT. Caspase-dependent and caspase-independent death pathways along with pro-survival pathways activation were explored. KEY RESULTS As previously reported for BDNF, olesoxime dose-dependently delayed CPT-induced cell death. Both compounds acted downstream of p53 activation preventing cytochrome c release and caspases activation. When caspase activation was blocked, both olesoxime and BDNF provided additional neuroprotective effect, potentially through the prevention of apoptosis-inducing factor release from mitochondria. While BDNF activates both the PI3K/Akt and the ERK pathway, olesoxime induced only a late activation of the ERK pathways, which did not seem to play a major role in its neuroprotection against CPT. Rather, our results favour preserved mitochondrial membrane integrity by olesoxime. CONCLUSIONS AND IMPLICATIONS Albeit different, olesoxime and BDNF mechanisms for neuroprotection converge to preserve mitochondrial function. These findings emphasize the importance of targeting the mitochondria in the process of neurodegeneration. Importantly olesoxime, by mimicking neurotrophin pro-survival activities without impacting PI3K/Akt and ERK signalling, may have greater therapeutic potential in many diseases where neurotrophins were considered as a therapeutic solution.
Collapse
|
37
|
Luo T, Yue R, Hu H, Zhou Z, Yiu KH, Zhang S, Xu L, Li K, Yu Z. PD150606 protects against ischemia/reperfusion injury by preventing μ-calpain-induced mitochondrial apoptosis. Arch Biochem Biophys 2015; 586:1-9. [PMID: 26091952 DOI: 10.1016/j.abb.2015.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/15/2015] [Accepted: 06/11/2015] [Indexed: 01/14/2023]
Abstract
Calpain plays an important role in myocardial ischemia/reperfusion (I/R) injury. PD150606, a nonpeptide, cell-permeable and noncompetitive calpain inhibitor, has been shown to have protective properties in ischemic disease. The aims of the present study were to investigate whether PD150606 could alleviate myocardial I/R injury and to examine the possible mechanisms involved. The I/R model was established in vivo in C57BL/6 mice and in vitro using neonatal mouse cardiomyocytes, respectively. To evaluate the protective effects of PD150606 on I/R injury, we measured the myocardial infarct area, apoptosis, and expression of cleaved caspase-3. We also investigated the underlying mechanisms by examining mitochondrial function as reflected by the ATP concentration, translocation of cytochrome c, dynamics of mPTP opening, and membrane potential (ΔΨm), coupled with calpain activity. Pretreatment with PD150606 significantly reduced the infarct area and apoptosis caused by I/R. PD150606 pretreatment also reduced mitochondrial dysfunction by inhibiting calpain activation. Moreover, we found that μ-calpain is the main contributor to I/R-induced calpain activation. Knockdown of μ-calpain with siRNA significantly reversed calpain activation, mitochondrial dysfunction, and cardiomyocyte apoptosis caused by I/R in vitro. Our results suggest that PD150606 may protect against I/R injury via preventing μ-calpain-induced mitochondrial apoptosis.
Collapse
Affiliation(s)
- Tao Luo
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China; Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Rongchuan Yue
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China
| | - Houxiang Hu
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China; Center for Medical Research, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China.
| | - Zhou Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, China
| | - Kai Hang Yiu
- Department of Medicine, University of Hong Kong, Hong Kong 999077, China
| | - Shuang Zhang
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China
| | - Lei Xu
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China
| | - Ke Li
- Department of Cardiology, North Sichuan Medical College First Affiliated Hospital, Nanchong 637000, Sichuan, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
38
|
Rasagiline and selegiline suppress calcium efflux from mitochondria by PK11195-induced opening of mitochondrial permeability transition pore: a novel anti-apoptotic function for neuroprotection. J Neural Transm (Vienna) 2015; 122:1399-407. [DOI: 10.1007/s00702-015-1398-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/03/2015] [Indexed: 12/23/2022]
|
39
|
Yu N, Wang S, Wang P, Li Y, Li S, Wang L, Chen H, Wang Y. The calcium uniporter regulates the permeability transition pore in isolated cortical mitochondria. Neural Regen Res 2015; 7:109-13. [PMID: 25767484 PMCID: PMC4354124 DOI: 10.3969/j.issn.1673-5374.2012.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/20/2011] [Indexed: 01/28/2023] Open
Abstract
To investigate the influence of the mitochondrial calcium uniporter on the mitochondrial permeability transition pore, the present study observed mitochondrial morphology in cortical neurons isolated from adult rats using transmission electron microscopy, and confirmed the morphology and activity of isolated mitochondria by detecting succinic dehydrogenase and monoamine oxidase, two mitochondrial enzymes. Isolated mitochondria were treated with either ruthenium red, an inhibitor of the uniporter, spermine, an activator of the uniporter, or in combination with cyclosporin A, an inhibitor of the mitochondrial permeability transition pore. Results showed that ruthenium red inhibited CaCl2-induced mitochondrial permeability transition pore opening, spermine enhanced opening, and cyclosporin A attenuated the effects of spermine. Results demonstrated that the mitochondrial calcium uniporter plays a role in regulating the mitochondrial permeability transition pore in mitochondria isolated from the rat brain cortex.
Collapse
Affiliation(s)
- Ning Yu
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Shilei Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Peng Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Yu Li
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Shuhong Li
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Li Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Hongbing Chen
- Cerebrovascular Disease Institute, Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Yanting Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| |
Collapse
|
40
|
Pagliaro P, Penna C. Redox signalling and cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1974-95. [PMID: 25303224 DOI: 10.1111/bph.12975] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022] Open
Abstract
The morbidity and mortality from coronary artery disease (CAD) remain significant worldwide. The treatment for acute myocardial infarction has improved over the past decades, including early reperfusion of culprit coronary arteries. Although it is mandatory to reperfuse the ischaemic territory as soon as possible, paradoxically this leads to additional myocardial injury, namely ischaemia/reperfusion (I/R) injury, in which redox stress plays a pivotal role and for which no effective therapy is currently available. In this review, we report evidence that the redox environment plays a pivotal role not only in I/R injury but also in cardioprotection. In fact, cardioprotective strategies, such as pre- and post-conditioning, result in a robust reduction in infarct size in animals and the role of redox signalling is of paramount importance in these conditioning strategies. Nitrosative signalling and cysteine redox modifications, such as S-nitrosation/S-nitrosylation, are also emerging as very important mechanisms in conditioning cardioprotection. The reasons for the switch from protective oxidative/nitrosative signalling to deleterious oxidative/nitrosative/nitrative stress are not fully understood. The complex regulation of this switch is, at least in part, responsible for the diminished or lack of cardioprotection induced by conditioning protocols observed in ageing animals and with co-morbidities as well as in humans. Therefore, it is important to understand at a mechanistic level the reasons for these differences before proposing a safe and useful transition of ischaemic or pharmacological conditioning. Indeed, more mechanistic novel therapeutic strategies are required to protect the heart from I/R injury and to improve clinical outcomes in patients with CAD.
Collapse
Affiliation(s)
- P Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, 10043, Orbassano, Turin, Italy
| | | |
Collapse
|
41
|
Martin LJ, Fancelli D, Wong M, Niedzwiecki M, Ballarini M, Plyte S, Chang Q. GNX-4728, a novel small molecule drug inhibitor of mitochondrial permeability transition, is therapeutic in a mouse model of amyotrophic lateral sclerosis. Front Cell Neurosci 2014; 8:433. [PMID: 25565966 PMCID: PMC4271619 DOI: 10.3389/fncel.2014.00433] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder in humans characterized by progressive degeneration of skeletal muscle and motor neurons in spinal cord, brainstem, and cerebral cortex causing skeletal muscle paralysis, respiratory insufficiency, and death. There are no cures or effective treatments for ALS. ALS can be inherited, but most cases are not associated with a family history of the disease. Mitochondria have been implicated in the pathogenesis but definitive proof of causal mechanisms is lacking. Identification of new clinically translatable disease mechanism-based molecular targets and small molecule drug candidates are needed for ALS patients. We tested the hypothesis in an animal model that drug modulation of the mitochondrial permeability transition pore (mPTP) is therapeutic in ALS. A prospective randomized placebo-controlled drug trial was done in a transgenic (tg) mouse model of ALS. We explored GNX-4728 as a therapeutic drug. GNX-4728 inhibits mPTP opening as evidenced by increased mitochondrial calcium retention capacity (CRC) both in vitro and in vivo. Chronic systemic treatment of G37R-human mutant superoxide dismutase-1 (hSOD1) tg mice with GNX-4728 resulted in major therapeutic benefits. GNX-4728 slowed disease progression and significantly improved motor function. The survival of ALS mice was increased significantly by GNX-4728 treatment as evidence by a nearly 2-fold extension of lifespan (360 days-750 days). GNX-4728 protected against motor neuron degeneration and mitochondrial degeneration, attenuated spinal cord inflammation, and preserved neuromuscular junction (NMJ) innervation in the diaphragm in ALS mice. This work demonstrates that a mPTP-acting drug has major disease-modifying efficacy in a preclinical mouse model of ALS and establishes mitochondrial calcium retention, and indirectly the mPTP, as targets for ALS drug development.
Collapse
Affiliation(s)
- Lee J. Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Pathobiology Graduate Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | | | - Margaret Wong
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Mark Niedzwiecki
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | | | | | - Qing Chang
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
42
|
Protective effect of shen-fu injection on neuronal mitochondrial function in a porcine model of prolonged cardiac arrest. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:523847. [PMID: 25505924 PMCID: PMC4258377 DOI: 10.1155/2014/523847] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 10/31/2014] [Indexed: 11/29/2022]
Abstract
Background. Shen-Fu injection (SFI) following cardiac arrest exhibits neurological effects, but its effect on neurological dysfunction is unclear. This study sought to investigate the protective effect of SFI on nerve cells in a porcine model of cardiac arrest. Methods. After eight minutes of untreated ventricular fibrillation (VF) and 2 minutes of basic life support, 24 pigs were randomized and divided into three cardiopulmonary resuscitation groups, which received central venous injection of either Shen-Fu (SFI group; 1.0 ml/kg), epinephrine (EP group; 0.02 mg/kg), or saline (SA group). Surviving pigs were sacrificed at 24 h after ROSC and brains were removed for analysis for morphologic changes of mitochondria by electron microscopy, for mitochondrial transmembrane potential (MTP) by flow cytometry, and for opening of the mitochondrial permeability transition pore (MPTP) by mitochondrial light scattering. Results. Compared with the EP and SA groups, SFI treatment reduced opening of MPTP, showing higher MMP. In addition, animals treated with SFI showed slight cerebral ultrastructure damage under the electron microscopy. Conclusion. Shen-Fu injection alleviated brain injury, improved neurological ultrastructure, stabilized membrane potential, and inhibited opening of MPTP. Therefore, SFI could significantly attenuate postresuscitation cerebral ischemia and reperfusion injury by modulating mitochondrial dysfunction of nerve cells.
Collapse
|
43
|
Liu Y, Li W, Hu L, Liu Y, Li B, Sun C, Zhang C, Zou L. Downregulation of nitric oxide by electroacupuncture against hypoxic‑ischemic brain damage in rats via nuclear factor‑κB/neuronal nitric oxide synthase. Mol Med Rep 2014; 11:837-42. [PMID: 25374015 PMCID: PMC4262503 DOI: 10.3892/mmr.2014.2879] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 09/09/2014] [Indexed: 01/13/2023] Open
Abstract
The present study aimed to investigate the role of nitric oxide (NO) against perinatal hypoxic‑ischemic brain damage (HIBD) in rats by electroacupuncture (EA) and to examine its potential neuroprotective mechanism. NO content, the number of positive cells, neuronal nitric oxide synthase (nNOS) and nuclear factor‑κB (NF‑κB) in rat cortex cells were determined. The results demonstrated that treatment with EA significantly downregulated the NO content in the cortex cells (*P<0.05, **P<0.01, compared with the control groups) and alleviated cell damage in the cortex of rats with HIBD. The activator, S‑adenosyl‑L‑methionine and the inhibitor, hydroxylamine of cystathionine‑β‑synthase (CBS), aggravated and remitted the hypoxic damage in the cortex cells, respectively. In addition, treatment with EA significantly downregulated the expression of nNOS and NF‑κB in the rat cortex cells (*P<0.05, **P<0.01, compared with the control groups). The results also indicated that treatment with EA downregulated the NO content of cortical cells against HIBD via the NF‑κB/nNOS pathway and further implied that the hydrogen sulfide/CBS system may be involved in the process. The present study provided a significant reference for the prevention and treatment of HIBD using the EA technique and also described a novel protective mechanism.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Pediatrics, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, P.R. China
| | - Weiguang Li
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLA, Beijing 100850, P.R. China
| | - Linyan Hu
- Department of Pediatrics, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, P.R. China
| | - Ying Liu
- Department of Pediatrics, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, P.R. China
| | - Baoquan Li
- Department of Pediatrics, 159th Hospital of Chinese People's Liberation Army, Zhumadian, Henan 463000, P.R. China
| | - Changqing Sun
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLA, Beijing 100850, P.R. China
| | - Chenggang Zhang
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center of PLA, Beijing 100850, P.R. China
| | - Liping Zou
- Department of Pediatrics, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, P.R. China
| |
Collapse
|
44
|
Cherry BH, Sumien N, Mallet RT. Neuronal injury from cardiac arrest: aging years in minutes. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9680. [PMID: 25104136 PMCID: PMC4150914 DOI: 10.1007/s11357-014-9680-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/26/2014] [Indexed: 06/03/2023]
Abstract
Cardiac arrest is a leading cause of death and permanent disability. Most victims succumb to the oxidative and inflammatory damage sustained during cardiac arrest/resuscitation, but even survivors typically battle long-term neurocognitive impairment. Although extensive research has delineated the complex mechanisms that culminate in neuronal damage and death, no effective treatments have been developed to interrupt these mechanisms. Of importance, many of these injury cascades are also active in the aging brain, where neurons and other cells are under persistent oxidative and inflammatory stress which eventually damages or kills the cells. In light of these similarities, it is reasonable to propose that the brain essentially ages the equivalent of several years within the few minutes taken to resuscitate a patient from cardiac arrest. Accordingly, cardiac arrest-resuscitation models may afford an opportunity to study the deleterious mechanisms underlying the aging process, on an accelerated time course. The aging and resuscitation fields both stand to gain pivotal insights from one another regarding the mechanisms of injury sustained during resuscitation from cardiac arrest and during aging. This synergism between the two fields could be harnessed to foster development of treatments to not only save lives but also to enhance the quality of life for the elderly.
Collapse
Affiliation(s)
- Brandon H Cherry
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA,
| | | | | |
Collapse
|
45
|
Kanngiesser M, Mair N, Lim HY, Zschiebsch K, Blees J, Häussler A, Brüne B, Ferreiròs N, Kress M, Tegeder I. Hypoxia-inducible factor 1 regulates heat and cold pain sensitivity and persistence. Antioxid Redox Signal 2014; 20:2555-71. [PMID: 24144405 DOI: 10.1089/ars.2013.5494] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS The present study assessed the functions of the transcription factor hypoxia-inducible factor (HIF) in sensory neurons in models of acute, inflammatory, ischemic, and neuropathic pain. The alpha subunit, HIF1α, was specifically deleted in neurons of the dorsal root ganglia by mating HIF1α(fl/fl) mice with SNScre mice. RESULTS SNS-HIF1α(-/-) mice were more sensitive to noxious heat and cold pain stimulation than were HIF1α(fl/fl) control mice. They also showed heightened first-phase nociceptive responses in the formalin and capsaicin tests with increased numbers of cFos-positive neurons in the dorsal horn, and intensified hyperalgesia in early phases after paw inflammation and hind limb ischemia/reperfusion. The behavioral cold and heat pain hypersensitivity was explained by increased calcium fluxes after transient receptor potential channel activation in primary sensory neurons of SNS-HIF1α(-/-) mice and lowered electrical activation thresholds of sensory fibers. SNS-HIF1α(-/-) mice however, developed less neuropathic pain after sciatic nerve injury, which was associated with an abrogation of HIF1-mediated gene up-regulation. INNOVATION The results suggest that HIF1α is protective in terms of acute heat and cold pain but in case of ongoing activation in injured neurons, it may promote the development of neuropathic pain. CONCLUSION The duality of HIF1 in pain regulation may have an impact on the side effects of drugs targeting HIF1, which are being developed, for example, as anticancer agents. Specifically, in patients with cancer neuropathy, however, temporary HIF1 inhibition might provide a welcome combination of growth and pain reduction.
Collapse
Affiliation(s)
- Maike Kanngiesser
- 1 Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Hospital Frankfurt , Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ma Q, Chen S, Hu Q, Feng H, Zhang JH, Tang J. Reply: To PMID 24273204. Ann Neurol 2014; 75:972-973. [PMID: 24805252 PMCID: PMC4145719 DOI: 10.1002/ana.24175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 05/02/2014] [Accepted: 05/02/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Qingyi Ma
- Department of Physiology and Pharmacology, Loma Linda University,
Loma Linda, California, USA
| | - Sheng Chen
- Department of Physiology and Pharmacology, Loma Linda University,
Loma Linda, California, USA
- Department of Neurosurgery, Second Affiliated Hospital, School of
Medicine, Zhejiang University, Zhejiang, China
| | - Qin Hu
- Department of Physiology and Pharmacology, Loma Linda University,
Loma Linda, California, USA
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military
Medical University, Chongqing, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University,
Loma Linda, California, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda,
California, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda,
California, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University,
Loma Linda, California, USA
| |
Collapse
|
47
|
Richter F, Gao F, Medvedeva V, Lee P, Bove N, Fleming SM, Michaud M, Lemesre V, Patassini S, De La Rosa K, Mulligan CK, Sioshansi PC, Zhu C, Coppola G, Bordet T, Pruss RM, Chesselet MF. Chronic administration of cholesterol oximes in mice increases transcription of cytoprotective genes and improves transcriptome alterations induced by alpha-synuclein overexpression in nigrostriatal dopaminergic neurons. Neurobiol Dis 2014; 69:263-75. [PMID: 24844147 DOI: 10.1016/j.nbd.2014.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/14/2022] Open
Abstract
Cholesterol-oximes TRO19622 and TRO40303 target outer mitochondrial membrane proteins and have beneficial effects in preclinical models of neurodegenerative diseases leading to their advancement to clinical trials. Dopaminergic neurons degenerate in Parkinson's disease (PD) and are prone to oxidative stress and mitochondrial dysfunction. In order to provide insights into the neuroprotective potential of TRO19622 and TRO40303 for dopaminergic neurons in vivo, we assessed their effects on gene expression in laser captured nigrostriatal dopaminergic neurons of wildtype mice and of mice that over-express alpha-synuclein, a protein involved in both familial and sporadic forms of PD (Thy1-aSyn mice). Young mice were fed the drugs in food pellets or a control diet from 1 to 4months of age, approximately 10months before the appearance of striatal dopamine loss in this model. Unbiased weighted gene co-expression network analysis (WGCNA) of transcriptional changes revealed effects of cholesterol oximes on transcripts related to mitochondria, cytoprotection and anti-oxidant response in wild-type and transgenic mice, including increased transcription of stress defense (e.g. Prdx1, Prdx2, Glrx2, Hspa9, Pink1, Drp1, Trak1) and dopamine-related (Th, Ddc, Gch1, Dat, Vmat2, Drd2, Chnr6a) genes. Even at this young age transgenic mice showed alterations in transcripts implicated in mitochondrial function and oxidative stress (e.g. Bcl-2, Bax, Casp3, Nos2), and both drugs normalized about 20% of these alterations. Young Thy1-aSyn mice exhibit motor deficits that differ from parkinsonism and are established before the onset of treatment; these deficits were not improved by cholesterol oximes. However, high doses of TRO40303 improved olfaction and produced the same effects as dopamine agonists on a challenging beam test, specifically an increase in footslips, an observation congruent with its effects on transcripts involved in dopamine synthesis. High doses of TRO19622 increased alpha-synuclein aggregates in the substantia nigra; this effect, not seen with TRO40303 was inconsistent and may represent a protective mechanism as in other neurodegenerative diseases. Overall, the results suggest that cholesterol oximes, while not improving early effects of alpha-synuclein overexpression on motor behavior or pathology, may ameliorate the function and resilience of dopaminergic neurons in vivo and support further studies of neuroprotection in models with dopaminergic cell loss.
Collapse
Affiliation(s)
- Franziska Richter
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Fuying Gao
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Vera Medvedeva
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Patrick Lee
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Nicholas Bove
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Sheila M Fleming
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Magali Michaud
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Vincent Lemesre
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Stefano Patassini
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Krystal De La Rosa
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Caitlin K Mulligan
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Pedrom C Sioshansi
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Chunni Zhu
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Giovanni Coppola
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA; Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Thierry Bordet
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Rebecca M Pruss
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Marie-Françoise Chesselet
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA.
| |
Collapse
|
48
|
Regulators of mitochondrial Ca2+ homeostasis in cerebral ischemia. Cell Tissue Res 2014; 357:395-405. [DOI: 10.1007/s00441-014-1807-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/10/2014] [Indexed: 02/06/2023]
|
49
|
Chen S, Feng H, Sherchan P, Klebe D, Zhao G, Sun X, Zhang J, Tang J, Zhang JH. Controversies and evolving new mechanisms in subarachnoid hemorrhage. Prog Neurobiol 2014; 115:64-91. [PMID: 24076160 PMCID: PMC3961493 DOI: 10.1016/j.pneurobio.2013.09.002] [Citation(s) in RCA: 270] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/07/2013] [Accepted: 09/12/2013] [Indexed: 12/13/2022]
Abstract
Despite decades of study, subarachnoid hemorrhage (SAH) continues to be a serious and significant health problem in the United States and worldwide. The mechanisms contributing to brain injury after SAH remain unclear. Traditionally, most in vivo research has heavily emphasized the basic mechanisms of SAH over the pathophysiological or morphological changes of delayed cerebral vasospasm after SAH. Unfortunately, the results of clinical trials based on this premise have mostly been disappointing, implicating some other pathophysiological factors, independent of vasospasm, as contributors to poor clinical outcomes. Delayed cerebral vasospasm is no longer the only culprit. In this review, we summarize recent data from both experimental and clinical studies of SAH and discuss the vast array of physiological dysfunctions following SAH that ultimately lead to cell death. Based on the progress in neurobiological understanding of SAH, the terms "early brain injury" and "delayed brain injury" are used according to the temporal progression of SAH-induced brain injury. Additionally, a new concept of the vasculo-neuronal-glia triad model for SAH study is highlighted and presents the challenges and opportunities of this model for future SAH applications.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Prativa Sherchan
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Damon Klebe
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Xiaochuan Sun
- Department of Neurosurgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
50
|
Viscomi MT, Molinari M. Remote neurodegeneration: multiple actors for one play. Mol Neurobiol 2014; 50:368-89. [PMID: 24442481 DOI: 10.1007/s12035-013-8629-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
Remote neurodegeneration significantly influences the clinical outcome in many central nervous system (CNS) pathologies, such as stroke, multiple sclerosis, and traumatic brain and spinal cord injuries. Because these processes develop days or months after injury, they are accompanied by a therapeutic window of opportunity. The complexity and clinical significance of remote damage is prompting many groups to examine the factors of remote degeneration. This research is providing insights into key unanswered questions, opening new avenues for innovative neuroprotective therapies. In this review, we evaluate data from various remote degeneration models to describe the complexity of the systems that are involved and the importance of their interactions in reducing damage and promoting recovery after brain lesions. Specifically, we recapitulate the current data on remote neuronal degeneration, focusing on molecular and cellular events, as studied in stroke and brain and spinal cord injury models. Remote damage is a multifactorial phenomenon in which many components become active in specific time frames. Days, weeks, or months after injury onset, the interplay between key effectors differentially affects neuronal survival and functional outcomes. In particular, we discuss apoptosis, inflammation, oxidative damage, and autophagy-all of which mediate remote degeneration at specific times. We also review current findings on the pharmacological manipulation of remote degeneration mechanisms in reducing damage and sustaining outcomes. These novel treatments differ from those that have been proposed to limit primary lesion site damage, representing new perspectives on neuroprotection.
Collapse
Affiliation(s)
- Maria Teresa Viscomi
- Experimental Neurorehabilitation Laboratory, Santa Lucia Foundation I.R.C.C.S., Via del Fosso di Fiorano 65, 00143, Rome, Italy,
| | | |
Collapse
|