1
|
Singh R, Kaur N, Choubey V, Dhingra N, Kaur T. Endoplasmic reticulum stress and its role in various neurodegenerative diseases. Brain Res 2024; 1826:148742. [PMID: 38159591 DOI: 10.1016/j.brainres.2023.148742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/07/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
The Endoplasmic reticulum (ER), a critical cellular organelle, maintains cellular homeostasis by regulating calcium levels and orchestrating essential functions such as protein synthesis, folding, and lipid production. A pivotal aspect of ER function is its role in protein quality control. When misfolded proteins accumulate within the ER due to factors like protein folding chaperone dysfunction, toxicity, oxidative stress, or inflammation, it triggers the Unfolded protein response (UPR). The UPR involves the activation of chaperones like calnexin, calreticulin, glucose-regulating protein 78 (GRP78), and Glucose-regulating protein 94 (GRP94), along with oxidoreductases like protein disulphide isomerases (PDIs). Cells employ the Endoplasmic reticulum-associated degradation (ERAD) mechanism to counteract protein misfolding. ERAD disruption causes the detachment of GRP78 from transmembrane proteins, initiating a cascade involving Inositol-requiring kinase/endoribonuclease 1 (IRE1), Activating transcription factor 6 (ATF6), and Protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathways. The accumulation and deposition of misfolded proteins within the cell are hallmarks of numerous neurodegenerative diseases. These aberrant proteins disrupt normal neuronal signalling and contribute to impaired cellular homeostasis, including oxidative stress and compromised protein degradation pathways. In essence, ER stress is defined as the cellular response to the accumulation of misfolded proteins in the endoplasmic reticulum, encompassing a series of signalling pathways and molecular events that aim to restore cellular homeostasis. This comprehensive review explores ER stress and its profound implications for the pathogenesis and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rimaljot Singh
- Department of Biophysics, Panjab University Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| | - Vinay Choubey
- Department of Pharmacology, University of Tartu, Ravila 19, 51014 Tartu, Estonia
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University Chandigarh, India.
| |
Collapse
|
2
|
Patel TA, Kevadiya BD, Bajwa N, Singh PA, Zheng H, Kirabo A, Li YL, Patel KP. Role of Nanoparticle-Conjugates and Nanotheranostics in Abrogating Oxidative Stress and Ameliorating Neuroinflammation. Antioxidants (Basel) 2023; 12:1877. [PMID: 37891956 PMCID: PMC10604131 DOI: 10.3390/antiox12101877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress is a deteriorating condition that arises due to an imbalance between the reactive oxygen species and the antioxidant system or defense of the body. The key reasons for the development of such conditions are malfunctioning of various cell organelles, such as mitochondria, endoplasmic reticulum, and Golgi complex, as well as physical and mental disturbances. The nervous system has a relatively high utilization of oxygen, thus making it particularly vulnerable to oxidative stress, which eventually leads to neuronal atrophy and death. This advances the development of neuroinflammation and neurodegeneration-associated disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, dementia, and other memory disorders. It is imperative to treat such conditions as early as possible before they worsen and progress to irreversible damage. Oxidative damage can be negated by two mechanisms: improving the cellular defense system or providing exogenous antioxidants. Natural antioxidants can normally handle such oxidative stress, but they have limited efficacy. The valuable features of nanoparticles and/or nanomaterials, in combination with antioxidant features, offer innovative nanotheranostic tools as potential therapeutic modalities. Hence, this review aims to represent novel therapeutic approaches like utilizing nanoparticles with antioxidant properties and nanotheranostics as delivery systems for potential therapeutic applications in various neuroinflammation- and neurodegeneration-associated disease conditions.
Collapse
Affiliation(s)
- Tapan A. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Neha Bajwa
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali 140413, Punjab, India; (N.B.); (P.A.S.)
| | - Preet Amol Singh
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali 140413, Punjab, India; (N.B.); (P.A.S.)
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| |
Collapse
|
3
|
Zhang X, Xia M, Wu Y, Zhang F. Branched-Chain Amino Acids Metabolism and Their Roles in Retinopathy: From Relevance to Mechanism. Nutrients 2023; 15:2161. [PMID: 37432261 DOI: 10.3390/nu15092161] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023] Open
Abstract
Retinopathy is one of the leading causes of irreversible blindness and vision loss worldwide. Imbalanced nutrients play important roles in the pathogenesis and pathophysiology of retinal diseases. Branched-Chain Amino Acids (BCAAs), as essential amino acids, perform a variety of biological functions, including protein synthesis, glucose metabolism, lipid metabolism, inflammation, and oxidative stress in metabolic tissues of diabetes and aging-related diseases. Recently, it has been shown that BCAAs are highly related to neuroprotection, oxidative stress, inflammatory and glutamate toxicity in the retina of retinopathy. Therefore, this review summarizes the alterations of BCAA levels in retinopathy, especially diabetic retinopathy and aging-related macular disease, and the genetics, functions, and mechanisms of BCAAs in the retina as well as other metabolic tissues for reference. All of these efforts aim to provide fundamental knowledge of BCAAs for further discoveries and research on retina health based on the sensing and signaling of essential amino acids.
Collapse
Affiliation(s)
- Xiaonan Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Liaoning Provence Key Laboratory of Genome Engineered Animal Models, National Center of Genetically Engineered Animal Models for International Research, Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian 116000, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Mengxue Xia
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Yingjie Wu
- Liaoning Provence Key Laboratory of Genome Engineered Animal Models, National Center of Genetically Engineered Animal Models for International Research, Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian 116000, China
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250021, China
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Fang Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| |
Collapse
|
4
|
Dong XY. Calcium Ion Channels in Saccharomyces cerevisiae. J Fungi (Basel) 2023; 9:jof9050524. [PMID: 37233235 DOI: 10.3390/jof9050524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Regulating calcium ion (Ca2+) channels to improve the cell cycle and metabolism is a promising technology, ensuring increased cell growth, differentiation, and/or productivity. In this regard, the composition and structure of Ca2+ channels play a vital role in controlling the gating states. In this review, Saccharomyces cerevisiae, as a model eukaryotic organism and an essential industrial microorganism, was used to discuss the effect of its type, composition, structure, and gating mechanism on the activity of Ca2+ channels. Furthermore, the advances in the application of Ca2+ channels in pharmacology, tissue engineering, and biochemical engineering are summarized, with a special focus on exploring the receptor site of Ca2+ channels for new drug design strategies and different therapeutic uses, targeting Ca2+ channels to produce functional replacement tissues, creating favorable conditions for tissue regeneration, and regulating Ca2+ channels to enhance biotransformation efficiency.
Collapse
Affiliation(s)
- Xiao-Yu Dong
- College of Life and Health, Dalian University, Dalian 116622, China
| |
Collapse
|
5
|
Kim TW. Targeting ER Stress with Saikosaponin A to Overcome Resistance under Radiation in Gastric Cancer Cells. Int J Mol Sci 2023; 24:ijms24065661. [PMID: 36982736 PMCID: PMC10052548 DOI: 10.3390/ijms24065661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Saikosaponin A is a triterpene saponin and a potentially bioactive compound derived from Bupleurum falcatum L. However, the molecular mechanisms and effects of saikosaponin A in gastric cancer remain unknown. In the present study, I evaluated the effects of saikosaponin A on cell death and endoplasmic reticulum stress via calcium and reactive oxygen species release. Targeting reactive oxygen species with diphenyleneiodonium and N-acetylcysteine inhibited cell death and protein kinase RNA-like ER kinase signaling pathway by down-regulating Nox4 and inducing glucose-regulated protein 78 exosomes. Furthermore, saikosaponin A caused a synergistic inhibitory effect of the epithelial mesenchymal transition phenomenon, indicating the reversible phenotype modulation by epithelial cells under radiation exposure in radiation-resistant gastric cancer cells. These results suggest that saikosaponin A-mediated calcium and reactive oxygen species-induced endoplasmic reticulum stress overcome radio-resistance and induce cell death under radiation in gastric cancer cells. Therefore, saikosaponin A in combination with radiation may be a potential strategy for gastric cancer therapy.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Gyeongbuk, Republic of Korea
| |
Collapse
|
6
|
Yoo YM, Joo SS. Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress. Int J Mol Sci 2023; 24:ijms24032381. [PMID: 36768703 PMCID: PMC9916953 DOI: 10.3390/ijms24032381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
As people age, their risks of developing degenerative diseases such as cancer, diabetes, Parkinson's Disease (PD), Alzheimer's Disease (AD), rheumatoid arthritis, and osteoporosis are generally increasing. Millions of people worldwide suffer from these diseases as they age. In most countries, neurodegenerative diseases are generally recognized as the number one cause afflicting the elderly. Endoplasmic reticulum (ER) stress has been suggested to be associated with some human neurological diseases, such as PD and AD. Melatonin, a neuroendocrine hormone mainly synthesized in the pineal gland, is involved in pleiotropically biological functions, including the control of the circadian rhythm, immune enhancement, and antioxidant, anti-aging, and anti-tumor effects. Although there are many papers on the prevention or suppression of diseases by melatonin, there are very few papers about the effects of melatonin on ER stress in neurons and neurodegenerative diseases. This paper aims to summarize and present the effects of melatonin reported so far, focusing on its effects on neurons and neurodegenerative diseases related to ER stress. Studies have shown that the primary target molecule of ER stress for melatonin is CHOP, and PERK and GRP78/BiP are the secondary target molecules. Therefore, melatonin is crucial in protecting neurons and treating neurodegeneration against ER stress.
Collapse
Affiliation(s)
- Yeong-Min Yoo
- East Coast Life Sciences Institute, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
- Correspondence: (Y.-M.Y.); (S.S.J.); Tel.: +82-10-2494-5309 (Y.-M.Y.); +82-33-640-2856 (S.S.J.); Fax: +82-33-640-2849 (Y.-M.Y. & S.S.J.)
| | - Seong Soo Joo
- Department of Marine Bioscience, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
- Correspondence: (Y.-M.Y.); (S.S.J.); Tel.: +82-10-2494-5309 (Y.-M.Y.); +82-33-640-2856 (S.S.J.); Fax: +82-33-640-2849 (Y.-M.Y. & S.S.J.)
| |
Collapse
|
7
|
Keylani K, Arbab Mojeni F, Khalaji A, Rasouli A, Aminzade D, Karimi MA, Sanaye PM, Khajevand N, Nemayandeh N, Poudineh M, Azizabadi Farahani M, Esfandiari MA, Haghshoar S, Kheirandish A, Amouei E, Abdi A, Azizinezhad A, Khani A, Deravi N. Endoplasmic reticulum as a target in cardiovascular diseases: Is there a role for flavonoids? Front Pharmacol 2023; 13:1027633. [PMID: 36703744 PMCID: PMC9871646 DOI: 10.3389/fphar.2022.1027633] [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: 09/21/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Flavonoids are found in natural health products and plant-based foods. The flavonoid molecules contain a 15-carbon skeleton with the particular structural construction of subclasses. The most flavonoid's critical subclasses with improved health properties are the catechins or flavonols (e.g., epigallocatechin 3-gallate from green tea), the flavones (e.g., apigenin from celery), the flavanones (e.g., naringenin from citrus), the flavanols (e.g., quercetin glycosides from berries, onion, and apples), the isoflavones (e.g., genistein from soya beans) and the anthocyanins (e.g., cyanidin-3-O-glucoside from berries). Scientific data conclusively demonstrates that frequent intake of efficient amounts of dietary flavonoids decreases chronic inflammation and the chance of oxidative stress expressing the pathogenesis of human diseases like cardiovascular diseases (CVDs). The endoplasmic reticulum (ER) is a critical organelle that plays a role in protein folding, post-transcriptional conversion, and transportation, which plays a critical part in maintaining cell homeostasis. Various stimuli can lead to the creation of unfolded or misfolded proteins in the endoplasmic reticulum and then arise in endoplasmic reticulum stress. Constant endoplasmic reticulum stress triggers unfolded protein response (UPR), which ultimately causes apoptosis. Research has shown that endoplasmic reticulum stress plays a critical part in the pathogenesis of several cardiovascular diseases, including diabetic cardiomyopathy, ischemic heart disease, heart failure, aortic aneurysm, and hypertension. Endoplasmic reticulum stress could be one of the crucial points in treating multiple cardiovascular diseases. In this review, we summarized findings on flavonoids' effects on the endoplasmic reticulum and their role in the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Kimia Keylani
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Arbab Mojeni
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Asma Rasouli
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Dlnya Aminzade
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nazanin Khajevand
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Nemayandeh
- Drug and Food Control Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Mohammad Ali Esfandiari
- Student Research Committee, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sepehr Haghshoar
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
| | - Ali Kheirandish
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Erfan Amouei
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Arash Azizinezhad
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Afshin Khani
- Department of Cardiovascular Disease, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,*Correspondence: Niloofar Deravi,
| |
Collapse
|
8
|
Integrated Profiles of Transcriptome and mRNA m6A Modification Reveal the Intestinal Cytotoxicity of Aflatoxin B1 on HCT116 Cells. Genes (Basel) 2022; 14:genes14010079. [PMID: 36672820 PMCID: PMC9858580 DOI: 10.3390/genes14010079] [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: 10/19/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Aflatoxin B1 (AFB1) is widely prevalent in foods and animal feeds and is one of the most toxic and carcinogenic aflatoxin subtypes. Existing studies have proved that the intestine is targeted by AFB1, and adverse organic effects have been observed. This study aimed to investigate the relationship between AFB1-induced intestinal toxicity and N6-methyladenosine (m6A) RNA methylation, which involves the post-transcriptional regulation of mRNA expression. The transcriptome-wide m6A methylome and transcriptome profiles in human intestinal cells treated with AFB1 are presented. Methylated RNA immunoprecipitation sequencing and mRNA sequencing were carried out to determine the distinctions in m6A methylation and different genes expressed in AFB1-induced intestinal toxicity. The results showed that there were 2289 overlapping genes of the differentially expressed mRNAs and differentially m6A-methylation-modified mRNAs. After enrichment of the signaling pathways and biological processes, these genes participated in the terms of the cell cycle, endoplasmic reticulum, tight junction, and mitophagy. In conclusion, the study demonstrated that AFB1-induced HCT116 injury was related to the disruptions to the levels of m6A methylation modifications of target genes and the abnormal expression of m6A regulators.
Collapse
|
9
|
Dai X, Yu F, Jiang Z, Dong B, Kong X. A fast fluorescent probe for tracing endoplasmic reticulum-located carboxylesterase in living cells. LUMINESCENCE 2022; 37:2067-2073. [PMID: 36200455 DOI: 10.1002/bio.4392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022]
Abstract
Carboxylesterase (CEs), mainly localized in endoplasmic reticulum (ER), are responsible for hydrolyzing compounds containing various ester bonds. They have been closely associated with drug metabolism and cellular homeostasis. Although some CE fluorescent probes have been developed, there are still a lack of probes that could target to the ER. Here, we developed a novel fluorescent probe CR with a specific ER anchor for monitoring CEs. In CR, p-toluenesulfonamide was chosen for precise ER targeting. A simple acetyl moiety was used as the CE response site and fluorescence modulation unit. During the spectral tests, CR displayed a fast response speed (within 10 s) towards CEs. In addition, it showed high sensitivity [limit of detection (LOD) = 5.1 × 10-3 U/ml] and high selectivity with CEs. In biological imaging, probe CR could especially locate in the ER in HepG2 cells. After cells were treated with orilistat, CR succeeded in monitoring the changes in the CEs. Importantly, CR also had the ability to trace the changes in CEs in a tunicamycin-induced ER stress model. Therefore, probe CR could be a powerful molecular tool for further investigating the functions of CEs in the ER.
Collapse
Affiliation(s)
- Xiaoyu Dai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Faqi Yu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Zekun Jiang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| | - Xiuqi Kong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong, China
| |
Collapse
|
10
|
Secondary Mechanisms of Neurotrauma: A Closer Look at the Evidence. Diseases 2022; 10:diseases10020030. [PMID: 35645251 PMCID: PMC9149951 DOI: 10.3390/diseases10020030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Traumatic central nervous system injury is a leading cause of neurological injury worldwide. While initial neuroresuscitative efforts are focused on ameliorating the effects of primary injury through patient stabilization, secondary injury in neurotrauma is a potential cause of cell death, oxidative stress, and neuroinflammation. These secondary injuries lack defined therapy. The major causes of secondary injury in neurotrauma include endoplasmic reticular stress, mitochondrial dysfunction, and the buildup of reactive oxygen or nitrogenous species. Stress to the endoplasmic reticulum in neurotrauma results in the overactivation of the unfolded protein response with subsequent cell apoptosis. Mitochondrial dysfunction can lead to the release of caspases and the buildup of reactive oxygen species; several characteristics make the central nervous system particularly susceptible to oxidative damage. Together, endoplasmic reticulum, mitochondrial, and oxidative stress can have detrimental consequences, beginning moments and lasting days to months after the primary injury. Understanding these causative pathways has led to the proposal of various potential treatment options.
Collapse
|
11
|
Nakka VP, Gogada R, Simhadri PK, Qadeer MA, Phanithi PB. Post-treatment with apocynin at a lower dose regulates the UPR branch of eIF2α and XBP-1 pathways after stroke. Brain Res Bull 2022; 182:1-11. [PMID: 35143926 DOI: 10.1016/j.brainresbull.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022]
Abstract
Stroke leads to disturbance in the physiology of the ER (Endoplasmic Reticulum) that triggers UPR (Unfolded Protein Response) pathways aimed to compensate neuronal cell damage. However, sustained UPR causes stressful conditions in the ER lumen forming abnormal protein aggregates. Stroke-induced oxidative stress also amalgamates with UPR to safeguard and ensure the proper functioning of brain cells. Thus we tested the effect of apocynin (a potent antioxidant) post-treatment in experimental stroke on the outcome of ER stress and UPR branch pathways. We administered a low dose of apocynin at 1 mg/kg (intraperitoneal) to adult Sprague-Dawley rats subjected to Middle Cerebral Artery Occlusion (MCAO) for two-time points. The first dose immediately after re-establishing the blood flow and another at 6 h of reperfusion. Apocynin post-treatment significantly reduced ROS (Reactive Oxygen Species) generation at an early reperfusion time point of 4 h. It preserved neuronal morphology, dendritic spine density, reduced protein aggregation, and brain damage after 24 h of reperfusion. Apocynin post-treatment regulates the two UPR branch pathways in our experimental paradigm. 1) Down-regulation of eIF2α (Eukaryotic Initiation Factor 2α) phosphorylation, and CHOP (C/EBP homologous protein) 2) by reducing the XBP-1 (X-Box binding Protein-1) mRNA splicing downstream to PERK (Protein Kinase RNA-Like ER Kinase) and IRE1α (Inositol Requiring Enzyme 1alpha) UPR pathways, respectively. Bioinformatics prediction showed that apocynin has binding sites for PERK (Protein Kinase RNA-Like ER Kinase) and IRE1α proteins. The amino acid residues interacting with apocynin were Cys891 and Gln889 (for PERK), and the amino acids Ser726, Arg722, and Ala719 (for IRE1α) lying within their activation loop. Overall, these studies indicate that apocynin post-treatment might regulate ER stress/UPR pathways and minimize stroke brain damage, thus having implications for developing newer strategies for stroke treatment.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India; Department of Biochemistry, Acharya Nagarjuna University, Andhra Pradesh 522510, India
| | - Raghu Gogada
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India; Department of Biochemistry and Plant Physiology, M.S. Swaminathan School of Agriculture, Centurion University of Technology and Management, Odisha 761211, India
| | - Praveen Kumar Simhadri
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India
| | | | - Prakash Babu Phanithi
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India.
| |
Collapse
|
12
|
Singh R, Kaur N, Dhingra N, Kaur T. Protein misfolding, ER Stress and Chaperones: An approach to develop chaperone-based therapeutics for Alzheimer's Disease. Int J Neurosci 2021:1-21. [PMID: 34402740 DOI: 10.1080/00207454.2021.1968859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disorder with complex etiology that eventually leads to dementia. The main culprit of AD is the extracellular deposition of β-amyloid (Aβ) and intracellular neurofibrillary tangles. The protein conformational change and protein misfolding are the key events of AD pathophysiology, therefore endoplasmic reticulum (ER) stress is an apparent consequence. ER, stress-induced unfolded protein response (UPR) mediators (viz. PERK, IRE1, and ATF6) have been reported widely in the AD brain. Considering these factors, preventing proteins misfolding or aggregation of tau or amyloidogenic proteins appears to be the best approach to halt its pathogenesis. Therefore, therapies through chemical and pharmacological chaperones came to light as an alternative for the treatment of AD. Diverse studies have demonstrated 4-phenylbutyric acid (4-PBA) as a potential therapeutic agent in AD. The current review outlined the mechanism of protein misfolding, different etiological features behind the progression of AD, the significance of ER stress in AD, and the potential therapeutic role of different chaperones to counter AD. The study also highlights the gaps in current knowledge of the chaperones-based therapeutic approach and the possibility of developing chaperones as a potential therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Rimaljot Singh
- Department of Biophysics, Panjab University Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| |
Collapse
|
13
|
Lee EJ, Chan P, Chea L, Kim K, Kaufman RJ, Lin JH. ATF6 is required for efficient rhodopsin clearance and retinal homeostasis in the P23H rho retinitis pigmentosa mouse model. Sci Rep 2021; 11:16356. [PMID: 34381136 PMCID: PMC8357971 DOI: 10.1038/s41598-021-95895-7] [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: 04/22/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Retinitis Pigmentosa (RP) is a blinding disease that arises from loss of rods and subsequently cones. The P23H rhodopsin knock-in (P23H-KI) mouse develops retinal degeneration that mirrors RP phenotype in patients carrying the orthologous variant. Previously, we found that the P23H rhodopsin protein was degraded in P23H-KI retinas, and the Unfolded Protein Response (UPR) promoted P23H rhodopsin degradation in heterologous cells in vitro. Here, we investigated the role of a UPR regulator gene, activating transcription factor 6 (Atf6), in rhodopsin protein homeostasis in heterozygous P23H rhodopsin (Rho+/P23H) mice. Significantly increased rhodopsin protein levels were found in Atf6-/-Rho+/P23H retinas compared to Atf6+/-Rho+/P23H retinas at early ages (~ P12), while rhodopsin mRNA levels were not different. The IRE1 pathway of the UPR was hyper-activated in young Atf6-/-Rho+/P23H retinas, and photoreceptor layer thickness was unchanged at this early age in Rho+/P23H mice lacking Atf6. By contrast, older Atf6-/-Rho+/P23H mice developed significantly increased retinal degeneration in comparison to Atf6+/-Rho+/P23H mice in all retinal layers, accompanied by reduced rhodopsin protein levels. Our findings demonstrate that Atf6 is required for efficient clearance of rhodopsin protein in rod photoreceptors expressing P23H rhodopsin, and that loss of Atf6 ultimately accelerates retinal degeneration in P23H-KI mice.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA.,USC ROSKI Eye Institute and Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Priscilla Chan
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Leon Chea
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Kyle Kim
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jonathan H Lin
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA. .,Department of Pathology, Stanford University, Palo Alto, CA, USA. .,VA Palo Alto Healthcare System, Palo Alto, CA, USA. .,School of Medicine, Stanford University, 300 Pasteur Dr. L235, Palo Alto, CA, 94305, USA.
| |
Collapse
|
14
|
Li J, Snyder EY, Tang FHF, Pasqualini R, Arap W, Sidman RL. Nna1 gene deficiency triggers Purkinje neuron death by tubulin hyperglutamylation and ER dysfunction. JCI Insight 2020; 5:136078. [PMID: 33004692 PMCID: PMC7566705 DOI: 10.1172/jci.insight.136078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Posttranslational glutamylation/deglutamylation balance in tubulins influences dendritic maturation and neuronal survival of cerebellar Purkinje neurons (PNs). PNs and some additional neuronal types degenerate in several spontaneous, independently occurring Purkinje cell degeneration (pcd) mice featuring mutant neuronal nuclear protein induced by axotomy (Nna1), a deglutamylase gene. This defective deglutamylase allows glutamylases to form hyperglutamylated tubulins. In pcd, all PNs die during postnatal “adolescence.” Neurons in some additional brain regions also die, mostly later than PNs. We show in laser capture microdissected single PNs, in cerebellar granule cell neuronal clusters, and in dissected hippocampus and substantia nigra that deglutamase mRNA and protein were virtually absent before pcd PNs degenerated, whereas glutaminase mRNA and protein remained normal. Hyperglutamylated microtubules and dimeric tubulins accumulated in pcd PNs and were involved in pcd PN death by glutamylase/deglutamylase imbalance. Importantly, treatment with a microtubule depolymerizer corrected the glutamylation/deglutamylation ratio, increasing PN survival. Further, before onset of neuronal death, pcd PNs displayed prominent basal polylisosomal masses rich in ER. We propose a “seesaw” metamorphic model summarizing mutant Nna1-induced tubulin hyperglutamylation, the pcd’s PN phenotype, and report that the neuronal disorder involved ER stress, unfolded protein response, and protein synthesis inhibition preceding PN death by apoptosis/necroptosis. Purkinje cell degeneration is due to ER stress, unfolded protein response, and protein synthesis inhibition preceding Purkinje neuron death by apoptosis/necroptosis.
Collapse
Affiliation(s)
- Jianxue Li
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Evan Y Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Fenny HF Tang
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Richard L Sidman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Gao Y, Luo C, Yao Y, Huang J, Fu H, Xia C, Ye G, Yu L, Han J, Fan Y, Tao L. IL-33 Alleviated Brain Damage via Anti-apoptosis, Endoplasmic Reticulum Stress, and Inflammation After Epilepsy. Front Neurosci 2020; 14:898. [PMID: 32982679 PMCID: PMC7487557 DOI: 10.3389/fnins.2020.00898] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-33 belongs to a novel chromatin-associated cytokine newly recognized by the IL-1 family, and its specific receptor is the orphan IL-1 receptor (ST2). Cumulative evidence suggests that IL-33 plays a crucial effect on the pathological changes and pathogenesis of central nervous system (CNS) diseases and injuries, such as recurrent neonatal seizures (RNS). However, the specific roles of IL-33 and its related molecular mechanisms in RNS remain confused. In the present study, we investigated the protein expression changes and co-localized cell types of IL-33 or ST2, as well as the effect of IL-33 on RNS-induced neurobehavioral defects, weight loss, and apoptosis. Moreover, an inhibitor of IL-33, anti-IL-33 was performed to further exploited underlying mechanisms. We found that administration of IL-33 up-regulated the expression levels of IL-33 and ST2, and increased the number of its co-localization with Olig-2-positive oligodendrocytes and NeuN-positive neurons at 72 h post-RNS. Noteworthily, RNS-induced neurobehavioral deficits, bodyweight loss, and spatial learning and memory impairment, as well as cell apoptosis, were reversed by IL-33 pretreatment. Additionally, the increase in IL-1β and TNF-α levels, up-regulation of ER stress, as well as a decrease in anti-apoptotic protein Bcl-2 and an increase in pro-apoptotic protein CC-3 induced by RNS are prevented by administration of IL-33. Moreover, IL-33 in combination with Anti-IL-33 significantly inverted the effects of IL-33 or Anti-IL-33 alone on apoptosis, ER stress, and inflammation. Collectively, these data suggest that IL-33 attenuates RNS-induced neurobehavioral disorders, bodyweight loss, and spatial learning and memory deficits, at least in part through mechanisms involved in inhibition of apoptosis, ER stress, and neuro-inflammation.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Forensic Science, Medical College of Soochow University, Suzhou, China.,Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.,Shanghai Key Laboratory of Forensic Medicine, Department of Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China
| | - Chengliang Luo
- Department of Forensic Science, Medical College of Soochow University, Suzhou, China
| | - Yi Yao
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Junjie Huang
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Huifang Fu
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Traditional Chinese Medicine Hospital, Nanjing, China
| | - Chongjian Xia
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Guanghua Ye
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Linsheng Yu
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Junge Han
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Yanyan Fan
- Department of Forensic Science, Wenzhou Medical University, Wenzhou, China.,The Forensic Center, Wenzhou Medical University, Wenzhou, China.,Center of Basic Medical Experiment, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Luyang Tao
- Department of Forensic Science, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Yang Y, Zhou Q, Gao A, Chen L, Li L. Endoplasmic reticulum stress and focused drug discovery in cardiovascular disease. Clin Chim Acta 2020; 504:125-137. [PMID: 32017925 DOI: 10.1016/j.cca.2020.01.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/28/2022]
Abstract
Endoplasmic reticulum (ER) is an intracellular membranous organelle involved in the synthesis, folding, maturation and post-translation modification of secretory and transmembrane proteins. Therefore, ER is closely related to the maintenance of intracellular homeostasis and the good balance between health and diseases. Endoplasmic reticulum stress (ERS) occurs when unfolded/misfolded proteins accumulate after disturbance of ER environment. In response to ERS, cells trigger an adaptive response called the Unfolded protein response (UPR), which helps cells cope with the stress. In recent years, a large number of studies show that ERS can aggravate cardiovascular diseases. ERS-related proteins expression in cardiovascular diseases is on the rise. Therefore, down-regulation of ERS is critical for alleviating symptoms of cardiovascular diseases, which may be used in the near future to treat cardiovascular diseases. This article reviews the relationship between ERS and cardiovascular diseases and drugs that inhibit ERS. Furthermore, we detail the role of ERS inhibitors in the treatment of cardiovascular disease. Drugs that inhibit ERS are considered as promising strategies for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yiyuan Yang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Qionglin Zhou
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Anbo Gao
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| |
Collapse
|
17
|
Godisela KK, Reddy SS, Reddy PY, Kumar CU, Reddy VS, Ayyagari R, Reddy GB. Role of sorbitol-mediated cellular stress response in obesity-associated retinal degeneration. Arch Biochem Biophys 2020; 679:108207. [PMID: 31760123 DOI: 10.1016/j.abb.2019.108207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Obesity is a global health problem associated with several diseases including ocular complications. Earlier we reported progressive retinal degeneration because of obesity in a spontaneous obese rat (WNIN/Ob) model. In the current study, we examined the molecular mechanisms leading to retinal degeneration in WNIN/Ob rat. METHODS Sorbitol was estimated by the fluorometric method in the retina of WNIN/Ob rats at different age (3-, 6- and 12- months), along with their respective lean rats. Immunoblotting was performed in the retina to assess the status of the insulin signaling pathway, ER stress and cellular stress (p38MAPK and ERK1/2). Human SK-N-SH cells were treated with 0.5 and 1.0 M sorbitol for 30 min to study insulin signaling, ER stress, and cellular stress. TUNEL assay was done to measure apoptosis. The retinal function in the rats was determined by electroretinogram. RESULTS A gradual but significantly higher intracellular sorbitol accumulation was observed in the retina of obese rats from 3- to 12-months. The cellular osmotic stress has activated the insulin signaling mechanism without activating AKT and also triggered ER stress. Both the stresses activated the ERK and p38MAPK signaling causing apoptosis in the retina leading to retinal degeneration. Retinal dysfunction was confirmed by altered scotopic and photopic electroretinogram responses. These in vivo results were mimicked in SK-N-SH cells when exposed to sorbitol in vitro. CONCLUSIONS These results suggest cellular stress due to sorbitol accumulation impairing the ER function, thereby leading to progressive retinal degeneration under obese conditions.
Collapse
Affiliation(s)
- Kishore K Godisela
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | | | - P Yadagiri Reddy
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - Ch Uday Kumar
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - V Sudhakar Reddy
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - Radha Ayyagari
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
18
|
Miao YY, Zhao W, Li GP, Gao Y, Du PF. Predicting Endoplasmic Reticulum Resident Proteins Using Auto-Cross Covariance Transformation With a U-Shaped Residue Weight-Transfer Function. Front Genet 2020; 10:1231. [PMID: 31921288 PMCID: PMC6932965 DOI: 10.3389/fgene.2019.01231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/06/2019] [Indexed: 11/13/2022] Open
Abstract
Background: The endoplasmic reticulum (ER) is an important organelle in eukaryotic cells. It is involved in many important biological processes, such as cell metabolism, protein synthesis, and post-translational modification. The proteins that reside within the ER are called ER-resident proteins. These proteins are closely related to the biological functions of the ER. The difference between the ER-resident proteins and other non-resident proteins should be carefully studied. Methods: We developed a support vector machine (SVM)-based method. We developed a U-shaped weight-transfer function and used it, along with the positional-specific physiochemical properties (PSPCP), to integrate together sequence order information, signaling peptides information, and evolutionary information. Result: Our method achieved over 86% accuracy in a jackknife test. We also achieved roughly 86% sensitivity and 67% specificity in an independent dataset test. Our method is capable of identifying ER-resident proteins.
Collapse
Affiliation(s)
- Yang-Yang Miao
- College of Intelligence and Computing, Tianjin University, Tianjin, China.,School of Chemical Engineering, Tianjin University, Tianjin, China
| | - Wei Zhao
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Guang-Ping Li
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Yang Gao
- School of Medicine, Nankai University, Tianjin, China
| | - Pu-Feng Du
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| |
Collapse
|
19
|
Li D, Paterson DJ. Pre-synaptic sympathetic calcium channels, cyclic nucleotide-coupled phosphodiesterases and cardiac excitability. Semin Cell Dev Biol 2019; 94:20-27. [PMID: 30658154 DOI: 10.1016/j.semcdb.2019.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
In sympathetic neurons innervating the heart, action potentials activate voltage-gated Ca2+ channels and evoke Ca2+ entry into presynaptic terminals triggering neurotransmitter release. Binding of transmitters to specific receptors stimulates signal transduction pathways that cause changes in cardiac function. The mechanisms contributing to presynaptic Ca2+ dynamics involve regulation of endogenous Ca2+ buffers, in particular the endoplasmic reticulum, mitochondria and cyclic nucleotide targeted pathways. The purpose of this review is to summarize and highlight recent findings about Ca2+ homeostasis in cardiac sympathetic neurons and how modulation of second messengers can drive neurotransmission and affect myocyte excitability in cardiovascular disease. Moreover, we discuss the underlying mechanism of abnormal intracellular Ca2+ homeostasis and signaling in these neurons, and speculate on the role of phosphodiesterases as a therapeutic target to restore normal autonomic transmission in disease states of overactivity.
Collapse
Affiliation(s)
- Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
20
|
Expressions of ATF6, XBP1, and GRP78 in normal tissue, atypical adenomatous hyperplasia, and adenocarcinoma of the lung. Hum Pathol 2018; 83:22-28. [PMID: 30121368 DOI: 10.1016/j.humpath.2018.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/09/2018] [Accepted: 08/12/2018] [Indexed: 01/18/2023]
Abstract
Little is known about the association between the atypical adenomatous hyperplasia (AAH)-adenocarcinoma in situ sequence of the lung and endoplasmic reticulum-stress responders such as ATF6, XBP1, and GRP78. Using stored tissues, we examined (i) the percentage of a splicing form (active form) of XBP1 messenger RNA in normal lung tissue (NLT) and adenocarcinoma (ACA; using reverse-transcription polymerase chain reaction); (ii) ATF6 and GRP78 protein expressions in NLT and ACA (using Western blotting analysis); (iii) ATF6, XBP1, and GRP78 protein expressions in NLT, AAH, and ACA, including some adenocarcinoma in situ (using immunohistochemistry); and (iv) the incidence of nuclear translocation of the 3 proteins in these lesions. The percentage of the splicing form of XBP1 messenger RNA showed a borderline difference between NLT and ACA (P = .068). In the Western blotting analysis, the nuclear fractions of ATF6 (including the active form) and GRP78 proteins were higher in ACA than in NLT. In the immunohistochemistry, the values obtained for the incidence of the nuclear translocation of ATF6, XBP1, and GRP78 proteins were as follows, respectively: 13.3%, 2.2%, and 0.5% in low-grade AAH; 37.9%, 2.3%, and 2.2% in high-grade AAH; and 47.2%, 10.6%, and 4.4% in ACA. A significant difference was detected between low-grade AAH and ACA for ATF6. In terms of nuclear translocation, high-grade AAH seemed intermediate between low-grade AAH and ACA. These results support endoplasmic reticulum-stress responses, such as nuclear translocation of these 3 proteins (including their active forms), being in parallel with the progression of the adenoma-carcinoma sequence in the lung.
Collapse
|
21
|
l-Glutamine Attenuates Apoptosis Induced by Endoplasmic Reticulum Stress by Activating the IRE1α-XBP1 Axis in IPEC-J2: A Novel Mechanism of l-Glutamine in Promoting Intestinal Health. Int J Mol Sci 2017; 18:ijms18122617. [PMID: 29206200 PMCID: PMC5751220 DOI: 10.3390/ijms18122617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/28/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022] Open
Abstract
Intestinal absorption and barrier malfunctions are associated with endoplasmic reticulum stress (ERS) in the intestine. We induced ERS by exposing the intestinal porcine epithelial cell line J2 (IPEC-J2) to tunicamycin (TUNI) to explore the potential of l-glutamine to reduce ERS-induced apoptosis. Our experiments demonstrated that exposing cells to TUNI results in spontaneous ERS and encourages the upregulation of glucose-regulated protein 78 (GRP78). Prolonged TUNI-induced ERS was found to increase apoptosis mediated by C/enhancer binding protein homologous protein (CHOP), accompanied by GRP78 downregulation. Treatment with l-glutamine was found to promote cell proliferation within the growth medium but to have little effect in basic Dulbecco’s modified Eagle medium. Finally, in the milieu of TUNI-induced ERS, l-glutamine was found to maintain a high level of GRP78, alleviate CHOP-mediated apoptosis and activate the inositol requiring enzyme 1α (IRE1α)-X-box binding protein 1 (XBP1) axis. A specific inhibitor of the IRE1α-XBP1 axis reversed the protective effect of l-glutamine by blocking the expression of IRE1α/XBP1s. We propose that the functional effect of l-glutamine on intestinal health may be partly due to its modulation of ERS and CHOP-mediated apoptosis.
Collapse
|
22
|
Ren G, Zhou Y, Liang G, Yang B, Yang M, King A, Wei H. General Anesthetics Regulate Autophagy via Modulating the Inositol 1,4,5-Trisphosphate Receptor: Implications for Dual Effects of Cytoprotection and Cytotoxicity. Sci Rep 2017; 7:12378. [PMID: 28959036 PMCID: PMC5620053 DOI: 10.1038/s41598-017-11607-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/23/2017] [Indexed: 02/07/2023] Open
Abstract
General anesthetics are both neuroprotective and neurotoxic with unclear mechanisms. General anesthetics may control cell survival via their effects on autophagy by activation of type 1 inositol triphosphate receptor (InsP3R-1). DT40 or SH-SY5Y cells with only or over 99% expression of InsP3R-1 were treated with isoflurane or propofol. Cell viability was determined by MTT reduction or LDH release assays. Apoptosis was determined by measuring Caspase-3 or by TUNEL assay. Autophagy activity was determined by measuring LC3 II and P62. We evaluated mitochondrial integrity using MitoTracker Green and cytosolic ATP levels. Fura2-AM was used to measure the concentrations of cytosolic calcium ([Ca2+]c). Propofol significantly increased peak and integrated calcium response (P < 0.001) in cells with InsP3R-1 but not in cells with triple knockout of InsP3R. Both propofol and isoflurane increased autophagy induction (P < 0.05) in an mTOR- and InsP3R- activity dependent manner. Short exposure to propofol adequately activated InsP3-1 to provide sufficient autophagy for cytoprotection, while prolonged exposure to propofol induced cell apoptosis via impairment of autophagy flux through over activation of InsP3-1. Propofol damaged mitochondria and decreased cytosolic ATP. The effects of general anesthetics on apoptosis and autophagy are closely integrated; both are caused by differential activation of the type 1 InsP3R.
Collapse
Affiliation(s)
- Gongyi Ren
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yachun Zhou
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bin Yang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Meirong Yang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Alexander King
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Kumar R, Kumari B, Kumar M. Prediction of endoplasmic reticulum resident proteins using fragmented amino acid composition and support vector machine. PeerJ 2017; 5:e3561. [PMID: 28890846 PMCID: PMC5588793 DOI: 10.7717/peerj.3561] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/20/2017] [Indexed: 12/15/2022] Open
Abstract
Background The endoplasmic reticulum plays an important role in many cellular processes, which includes protein synthesis, folding and post-translational processing of newly synthesized proteins. It is also the site for quality control of misfolded proteins and entry point of extracellular proteins to the secretory pathway. Hence at any given point of time, endoplasmic reticulum contains two different cohorts of proteins, (i) proteins involved in endoplasmic reticulum-specific function, which reside in the lumen of the endoplasmic reticulum, called as endoplasmic reticulum resident proteins and (ii) proteins which are in process of moving to the extracellular space. Thus, endoplasmic reticulum resident proteins must somehow be distinguished from newly synthesized secretory proteins, which pass through the endoplasmic reticulum on their way out of the cell. Approximately only 50% of the proteins used in this study as training data had endoplasmic reticulum retention signal, which shows that these signals are not essentially present in all endoplasmic reticulum resident proteins. This also strongly indicates the role of additional factors in retention of endoplasmic reticulum-specific proteins inside the endoplasmic reticulum. Methods This is a support vector machine based method, where we had used different forms of protein features as inputs for support vector machine to develop the prediction models. During training leave-one-out approach of cross-validation was used. Maximum performance was obtained with a combination of amino acid compositions of different part of proteins. Results In this study, we have reported a novel support vector machine based method for predicting endoplasmic reticulum resident proteins, named as ERPred. During training we achieved a maximum accuracy of 81.42% with leave-one-out approach of cross-validation. When evaluated on independent dataset, ERPred did prediction with sensitivity of 72.31% and specificity of 83.69%. We have also annotated six different proteomes to predict the candidate endoplasmic reticulum resident proteins in them. A webserver, ERPred, was developed to make the method available to the scientific community, which can be accessed at http://proteininformatics.org/mkumar/erpred/index.html. Discussion We found that out of 124 proteins of the training dataset, only 66 proteins had endoplasmic reticulum retention signals, which shows that these signals are not an absolute necessity for endoplasmic reticulum resident proteins to remain inside the endoplasmic reticulum. This observation also strongly indicates the role of additional factors in retention of proteins inside the endoplasmic reticulum. Our proposed predictor, ERPred, is a signal independent tool. It is tuned for the prediction of endoplasmic reticulum resident proteins, even if the query protein does not contain specific ER-retention signal.
Collapse
Affiliation(s)
- Ravindra Kumar
- Department of Biophysics, University of Delhi South Campus, New Delhi, India.,Current affiliation: Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, Israel
| | - Bandana Kumari
- Department of Biophysics, University of Delhi South Campus, New Delhi, India
| | - Manish Kumar
- Department of Biophysics, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
24
|
Sorsby fundus dystrophy - A review of pathology and disease mechanisms. Exp Eye Res 2017; 165:35-46. [PMID: 28847738 DOI: 10.1016/j.exer.2017.08.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 01/29/2023]
Abstract
Sorsby fundus dystrophy (SFD) is an autosomal dominant macular dystrophy with an estimated prevalence of 1 in 220,000 and an onset of disease around the 4th to 6th decade of life. Similar to age-related macular degeneration (AMD), ophthalmoscopy reveals accumulation of protein/lipid deposits under the retinal pigment epithelium (RPE), referred to as drusen, in the eyes of patients with SFD. SFD is caused by variants in the gene for tissue inhibitor of metalloproteinases-3 (TIMP3), which has been found in drusen-like deposits of SFD patients. TIMP3 is constitutively expressed by RPE cells and, in healthy eyes, resides in Bruch's membrane. Most SFD-associated TIMP3 variants involve the gain or loss of a cysteine residue. This suggests the protein aberrantly forms intermolecular disulphide bonds, resulting in the formation of TIMP3 dimers. It has been demonstrated that SFD-associated TIMP3 variants are more resistant to turnover, which is thought to be a result of dimerisation and thought to explain the accumulation of TIMP3 in drusen-like deposits at the level of Bruch's membrane. An important function of TIMP3 within the outer retina is to regulate the thickness of Bruch's membrane. TIMP3 performs this function by inhibiting the activity of matrix metalloproteinases (MMPs), which have the function of catalysing breakdown of the extracellular matrix. TIMP3 has an additional function to inhibit vascular endothelial growth factor (VEGF) signalling and thereby to inhibit angiogenesis. However, it is unclear whether SFD-associated TIMP3 variant proteins retain these functions. In this review, we discuss the current understanding of the potential mechanisms underlying development of SFD and summarise all known SFD-associated TIMP3 variants. Cell culture models provide an invaluable way to study disease and identify potential treatments. These allow a greater understanding of RPE physiology and pathophysiology, including the ability to study the blood-retinal barrier as well as other RPE functions such as phagocytosis of photoreceptor outer segments. This review describes some examples of such recent in vitro studies and how they might provide new insights into degenerative diseases like SFD. Thus far, most studies on SFD have been performed using ARPE-19 cells or other, less suitable, cell-types. Now, induced pluripotent stem cell (iPSC) technologies allow the possibility to non-invasively collect somatic cells, such as dermal fibroblast cells and reprogram those to produce iPSCs. Subsequent differentiation of iPSCs can generate patient-derived RPE cells that carry the same disease-associated variant as RPE cells in the eyes of the patient. Use of these patient-derived RPE cells in novel cell culture systems should increase our understanding of how SFD and similar macular dystrophies develop.
Collapse
|
25
|
Development of a fluorescent reporter system for monitoring ER stress in Chinese hamster ovary cells and its application for therapeutic protein production. PLoS One 2017; 12:e0183694. [PMID: 28832690 PMCID: PMC5568292 DOI: 10.1371/journal.pone.0183694] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/09/2017] [Indexed: 12/31/2022] Open
Abstract
Mammalian cell expression systems have become a workhorse for the production of biotherapeutic proteins. As such, there is an ever increasing demand for higher productivity from these expression platforms to reduce manufacturing costs. While great advances have been made in the optimization of culture conditions and cell line selection to improve productivity, protein mis-folding remains a common limitation to high levels of production of therapeutic proteins. Accumulation of mis- and unfolded protein in the endoplasmic reticulum (ER) causes ER stress and initiates the unfolded protein response (UPR) that results in an activation of protein folding machinery, translation attenuation in an effort to proper folding of the newly synthesized peptides or may even lead to apoptosis if the correct folding is not restored. As a result, UPR associated apoptosis often results in lower protein expression. To better understand the molecular mechanisms in these pathways, we developed a reporter construct that detects Inositol-requiring enzyme 1 (IRE1)-alpha mediated splicing of X-box binding protein 1 (XBP1) to monitor the course of UPR activation in cell lines expressing monoclonal antibodies. Using this reporter we observed a clear activation of UPR in cells treated with known ER stress causing pharmacological agents, such as Tunicamycin (Tm) and Thapsigargin (Tg), as well as in stable IgG expressing cells during fed-batch cultures. Furthermore, we developed a stress metric that we term as ER stress index (ERSI) to gauge basal ER stress in cells which we used as a predictive tool for isolation of high IgG expressing cell lines. This reporter system, with its ability to monitor the stress involved in recombinant protein expression, has utility to assist in devising engineering strategies for improved production of biotherapeutic drugs.
Collapse
|
26
|
Jiang M, Yu S, Yu Z, Sheng H, Li Y, Liu S, Warner DS, Paschen W, Yang W. XBP1 (X-Box-Binding Protein-1)-Dependent O-GlcNAcylation Is Neuroprotective in Ischemic Stroke in Young Mice and Its Impairment in Aged Mice Is Rescued by Thiamet-G. Stroke 2017; 48:1646-1654. [PMID: 28487326 DOI: 10.1161/strokeaha.117.016579] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 03/09/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Impaired protein homeostasis induced by endoplasmic reticulum dysfunction is a key feature of a variety of age-related brain diseases including stroke. To restore endoplasmic reticulum function impaired by stress, the unfolded protein response is activated. A key unfolded protein response prosurvival pathway is controlled by the endoplasmic reticulum stress sensor (inositol-requiring enzyme-1), XBP1 (downstream X-box-binding protein-1), and O-GlcNAc (O-linked β-N-acetylglucosamine) modification of proteins (O-GlcNAcylation). Stroke impairs endoplasmic reticulum function, which activates unfolded protein response. The rationale of this study was to explore the potentials of the IRE1/XBP1/O-GlcNAc axis as a target for neuroprotection in ischemic stroke. METHODS Mice with Xbp1 loss and gain of function in neurons were generated. Stroke was induced by transient or permanent occlusion of the middle cerebral artery in young and aged mice. Thiamet-G was used to increase O-GlcNAcylation. RESULTS Deletion of Xbp1 worsened outcome after transient and permanent middle cerebral artery occlusion. After stroke, O-GlcNAcylation was activated in neurons of the stroke penumbra in young mice, which was largely Xbp1 dependent. This activation of O-GlcNAcylation was impaired in aged mice. Pharmacological increase of O-GlcNAcylation before or after stroke improved outcome in both young and aged mice. CONCLUSIONS Our study indicates a critical role for the IRE1/XBP1 unfolded protein response branch in stroke outcome. O-GlcNAcylation is a prosurvival pathway that is activated in the stroke penumbra in young mice but impaired in aged mice. Boosting prosurvival pathways to counterbalance the age-related decline in the brain's self-healing capacity could be a promising strategy to improve ischemic stroke outcome in aged brains.
Collapse
Affiliation(s)
- Meng Jiang
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Shu Yu
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Zhui Yu
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Huaxin Sheng
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Ying Li
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Shuai Liu
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - David S Warner
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.)
| | - Wulf Paschen
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.).
| | - Wei Yang
- From the Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC (M.J., S.Y., Z.Y., H.S., Y.L., S.L., D.S.W., W.P., W.Y.); Department of Anesthesiology (M.J.) and Department of Critical Care Medicine (Z.Y.), Renmin Hospital of Wuhan University, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Collaborative Innovation Center of Neuroregeneration, Nantong University, China (S.Y.); and Department of Cardiology, The Fifth Central Hospital of Tianjin, China (Y.L.).
| |
Collapse
|
27
|
Yu Z, Sheng H, Liu S, Zhao S, Glembotski CC, Warner DS, Paschen W, Yang W. Activation of the ATF6 branch of the unfolded protein response in neurons improves stroke outcome. J Cereb Blood Flow Metab 2017; 37:1069-1079. [PMID: 27217380 PMCID: PMC5363481 DOI: 10.1177/0271678x16650218] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Impaired function of the endoplasmic reticulum (ER stress) is a hallmark of many human diseases including stroke. To restore ER function in stressed cells, the unfolded protein response (UPR) is induced, which activates 3 ER stress sensor proteins including activating transcription factor 6 (ATF6). ATF6 is then cleaved by proteases to form the short-form ATF6 (sATF6), a transcription factor. To determine the extent to which activation of the ATF6 UPR branch defines the fate and function of neurons after stroke, we generated a conditional and tamoxifen-inducible sATF6 knock-in mouse. To express sATF6 in forebrain neurons, we crossed our sATF6 knock-in mouse line with Emx1-Cre mice to generate ATF6-KI mice. After the ATF6 branch was activated in ATF6-KI mice with tamoxifen, mice were subjected to transient middle cerebral artery occlusion. Forced activation of the ATF6 UPR branch reduced infarct volume and improved functional outcome at 24 h after stroke. Increased autophagic activity at early reperfusion time after stroke may contribute to the ATF6-mediated neuroprotection. We concluded that the ATF6 UPR branch is crucial to ischemic stroke outcome. Therefore, boosting UPR pro-survival pathways may be a promising therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Zhui Yu
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,2 Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huaxin Sheng
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Shuai Liu
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Shengli Zhao
- 3 Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | | | - David S Warner
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wulf Paschen
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- 1 Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
28
|
Tong J, Okutani F, Murata Y, Taniguchi M, Namba T, Wang YJ, Kaba H. Tunicamycin impairs olfactory learning and synaptic plasticity in the olfactory bulb. Neuroscience 2017; 344:371-379. [PMID: 28087337 DOI: 10.1016/j.neuroscience.2017.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/25/2016] [Accepted: 01/02/2017] [Indexed: 01/05/2023]
Abstract
Tunicamycin (TM) induces endoplasmic reticulum (ER) stress and inhibits N-glycosylation in cells. ER stress is associated with neuronal death in neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease, and most patients complain of the impairment of olfactory recognition. Here we examined the effects of TM on aversive olfactory learning and the underlying synaptic plasticity in the main olfactory bulb (MOB). Behavioral experiments demonstrated that the intrabulbar infusion of TM disabled aversive olfactory learning without affecting short-term memory. Histological analyses revealed that TM infusion upregulated C/EBP homologous protein (CHOP), a marker of ER stress, in the mitral and granule cell layers of MOB. Electrophysiological data indicated that TM inhibited tetanus-induced long-term potentiation (LTP) at the dendrodendritic excitatory synapse from mitral to granule cells. A low dose of TM (250nM) abolished the late phase of LTP, and a high dose (1μM) inhibited the early and late phases of LTP. Further, high-dose, but not low-dose, TM reduced the paired-pulse facilitation ratio, suggesting that the inhibitory effects of TM on LTP are partially mediated through the presynaptic machinery. Thus, our results support the hypothesis that TM-induced ER stress impairs olfactory learning by inhibiting synaptic plasticity via presynaptic and postsynaptic mechanisms in MOB.
Collapse
Affiliation(s)
- Jia Tong
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | - Fumino Okutani
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan; Department of Occupational Health, Kochi Medical School, Nankoku, Kochi 783-8505, Japan.
| | - Yoshihiro Murata
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | - Mutsuo Taniguchi
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | - Toshiharu Namba
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | - Yu-Jie Wang
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | - Hideto Kaba
- Department of Physiology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| |
Collapse
|
29
|
Yang W, Paschen W. Unfolded protein response in brain ischemia: A timely update. J Cereb Blood Flow Metab 2016; 36:2044-2050. [PMID: 27733676 PMCID: PMC5363674 DOI: 10.1177/0271678x16674488] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/23/2016] [Indexed: 01/13/2023]
Abstract
Folding and processing newly synthesized proteins are vital functions of the endoplasmic reticulum that are sensitive to a variety of stress conditions. The unfolded protein response is activated to restore endoplasmic reticulum function impaired by stress. While we know that brain ischemia impairs endoplasmic reticulum function, the role of unfolded protein response activation in post-ischemic recovery of neurologic function is only beginning to emerge. Here, we summarize what is known about endoplasmic reticulum stress and unfolded protein response in brain ischemia and discuss recent findings from myocardial ischemia studies that could help to advance research on endoplasmic reticulum stress and unfolded protein response in brain ischemia.
Collapse
Affiliation(s)
- Wei Yang
- Department of Anesthesiology, Duke University Medical Center, Durham, USA
| | - Wulf Paschen
- Department of Anesthesiology, Duke University Medical Center, Durham, USA.,Department of Neurobiology, Duke University Medical Center, Durham, USA
| |
Collapse
|
30
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
31
|
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder that has no known cure, nor is there a clear mechanistic understanding of the disease process itself. Although amyloid plaques, neurofibrillary tangles, and cognitive decline are late-stage markers of the disease, it is unclear how they are initially generated, and if they represent a cause, effect, or end phase in the pathology process. Recent studies in AD models have identified marked dysregulations in calcium signaling and related downstream pathways, which occur long before the diagnostic histopathological or cognitive changes. Under normal conditions, intracellular calcium signals are coupled to effectors that maintain a healthy physiological state. Consequently, sustained up-regulation of calcium may have pathophysiological consequences. Indeed, upon reviewing the current body of literature, increased calcium levels are functionally linked to the major features and risk factors of AD: ApoE4 expression, presenilin and APP mutations, beta amyloid plaques, hyperphosphorylation of tau, apoptosis, and synaptic dysfunction. In turn, the histopathological features of AD, once formed, are capable of further increasing calcium levels, leading to a rapid feed-forward acceleration once the disease process has taken hold. The views proposed here consider that AD pathogenesis reflects long-term calcium dysregulations that ultimately serve an enabling role in the disease process. Therefore, “Calcinists” do not necessarily reject βAptist or Tauist doctrine, but rather believe that their genesis is associated with earlier calcium signaling dysregulations. NEUROSCIENTIST 13(5):546—559, 2007.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL 60064, USA.
| |
Collapse
|
32
|
Upadhyay A, Amanullah A, Chhangani D, Mishra R, Mishra A. Selective multifaceted E3 ubiquitin ligases barricade extreme defense: Potential therapeutic targets for neurodegeneration and ageing. Ageing Res Rev 2015; 24:138-59. [PMID: 26247845 DOI: 10.1016/j.arr.2015.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/24/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022]
Abstract
Efficient and regular performance of Ubiquitin Proteasome System and Autophagy continuously eliminate deleterious accumulation of nonnative protiens. In cellular quality control system, E3 ubiquitin ligases are significant employees for defense mechanism against abnormal toxic proteins. Few findings indicate that lack of functions of E3 ubiquitin ligases can be a causative factor of neurodevelopmental disorders, neurodegeneration, cancer and ageing. However, the detailed molecular pathomechanism implying E3 ubiquitin ligases in cellular functions in multifactorial disease conditions are not well understood. This article systematically represents the unique characteristics, molecular nature, and recent developments in the knowledge of neurobiological functions of few crucial E3 ubiquitin ligases. Here, we review recent literature on the roles of E6-AP, HRD1 and ITCH E3 ubiquitin ligases in the neuro-pathobiological mechanisms, with precise focus on the processes of neurodegeneration, and thereby propose new lines of potential targets for therapeutic interventions.
Collapse
|
33
|
Mahdi AA, Rizvi SHM, Parveen A. Role of Endoplasmic Reticulum Stress and Unfolded Protein Responses in Health and Diseases. Indian J Clin Biochem 2015; 31:127-37. [PMID: 27069320 DOI: 10.1007/s12291-015-0502-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/12/2015] [Indexed: 12/24/2022]
Abstract
Endoplasmic reticulum (ER) is the site of protein synthesis, protein folding, maintainance of calcium homeostasis, synthesis of lipids and sterols. Genetic or environmental insults can alter its function generating ER stress. ER senses stress mainly by three stress sensor pathways, namely protein kinase R-like endoplasmic reticulum kinase-eukaryotic translation-initiation factor 2α, inositol-requiring enzyme 1α-X-box-binding protein 1 and activating transcription factor 6-CREBH, which induce unfolded protein responses (UPR) after the recognition of stress. Recent studies have demonstrated that ER stress and UPR signaling are involved in cancer, metabolic disorders, inflammatory diseases, osteoporosis and neurodegenerative diseases. However, the precise knowledge regarding involvement of ER stress in different disease processes is still debatable. Here we discuss the possible role of ER stress in various disorders on the basis of existing literature. An attempt has also been made to highlight the present knowledge of this field which may help to elucidate and conjure basic mechanisms and novel insights into disease processes which could assist in devising better future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Abbas Ali Mahdi
- Department of Biochemistry, King George's Medical University, Lucknow, 226003 Uttar Pradesh India
| | | | - Arshiya Parveen
- Department of Biochemistry, King George's Medical University, Lucknow, 226003 Uttar Pradesh India
| |
Collapse
|
34
|
XIAO BING, YU BO, LIU DONGJUAN, HAN FANG, SHI YUXIU. Single prolonged stress induces dysfunction of endoplasmic reticulum in a rat model of post-traumatic stress disorder. Mol Med Rep 2015; 12:2015-20. [DOI: 10.3892/mmr.2015.3590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 03/16/2015] [Indexed: 11/06/2022] Open
|
35
|
Tan H, Zou W, Jiang J, Tian Y, Xiao Z, Bi L, Zeng H, Tang X. Disturbance of hippocampal H2S generation contributes to CUMS-induced depression-like behavior: involvement in endoplasmic reticulum stress of hippocampus. Acta Biochim Biophys Sin (Shanghai) 2015; 47:285-91. [PMID: 25736403 DOI: 10.1093/abbs/gmv009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The chronic unpredictable mild stress (CUMS) model is a widely used experimental model of depression. Exogenous stress-induced neuronal cell death in the hippocampus is closely associated with the pathogenesis of depression. Excessive and prolonged endoplasmic reticulum (ER) stress triggers cell death. Hydrogen sulfide (H2S), the third endogenous signaling gasotransmitter, plays an important role in brain functions as a neuromodulator and a neuroprotectant. We hypothesized that the disturbance of endogenous H2S generation and ER stress in the hippocampus might be involved in CUMS-induced depression-like behaviors. Thus, the present study focused on whether CUMS disturbs the generation of endogenous H2S and up-regulates ER stress in the hippocampus and whether exogenous H2S prevents CUMS-induced depressive-like behaviors. Results showed that CUMS-treated rats exhibit depression-like behavior and hippocampal ER stress responses including up-regulated levels of glucose-regulated protein 78, CCAAT/enhancer binding protein homologous protein, and cleaved caspase-12 expression, while the endogenous generation of H2S in the hippocampus is suppressed in CUMS-treated rats. Furthermore, exogenous H2S prevents CUMS-induced depression-like behavior. These data indicated that CUMS-induced depression-like behaviors are related to the disturbance of endogenous H2S generation and ER stress in the hippocampus and suggested that endogenous H2S and ER stress are novel treatment targets of depression.
Collapse
Affiliation(s)
- Huiying Tan
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Wei Zou
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Jiamei Jiang
- Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Ying Tian
- Department of Biochemistry, University of South China, Hengyang 421001, China
| | - Zhifang Xiao
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China
| | - Lili Bi
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China
| | - Haiying Zeng
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Xiaoqing Tang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China Department of Physiology and Institute of Neuroscience, Medical College, University of South China, Hengyang 421001, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| |
Collapse
|
36
|
Boriushkin E, Wang JJ, Li J, Jing G, Seigel GM, Zhang SX. Identification of p58IPK as a novel neuroprotective factor for retinal neurons. Invest Ophthalmol Vis Sci 2015; 56:1374-86. [PMID: 25655802 DOI: 10.1167/iovs.14-15196] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Endoplasmic reticulum (ER)-resident chaperone protein p58(IPK) plays a vital role in regulation of protein folding and biosynthesis. The goal of this study was to examine the role of p58(IPK) in retinal neuronal cells under normal and stressed conditions. METHODS Retinal expression of p58(IPK), retinal morphology, apoptosis, ER stress, and apoptotic gene expression were examined in p58(IPK) knockout (KO) and/or wild-type (WT) mice with or without intravitreal injection of N-methyl-D-aspartic acid (NMDA). In in vitro experiments, differentiated R28 retinal neuronal cells transduced with adenovirus encoding p58(IPK) (Ad-p58(IPK)) or control virus (Ad-LacZ) were exposed to tunicamycin (TM) or hydrogen peroxide (H2O2). Levels of ER stress, apoptosis, and cell survival were evaluated. RESULTS Chaperone protein p58(IPK) is expressed predominantly in retinal ganglion cells (RGC), inner retinal neurons, and the photoreceptor inner segments. Mice lacking p58(IPK) exhibited increased CHOP expression and loss of RGCs with aging (8-10 months). Intravitreal injection of NMDA induced retinal ER stress and increased p58(IPK) expression in WT mice; this resulted in greater ER stress and enhanced RGC apoptosis in p58(IPK) KO mice. In cultured R28 cells, overexpression of p58(IPK) significantly reduced eIF2α phosphorylation, decreased CHOP expression, and alleviated the activation of caspase-3 and PARP. Overexpression of p58(IPK) also protected against oxidative and ER stress-induced cell apoptosis. Furthermore, p58(IPK) downregulated the proapoptotic gene Bax and upregulated the antiapoptotic gene Bcl-2 expression in stressed R28 cells. CONCLUSIONS Our study has demonstrated a protective role of p58(IPK) in retinal neurons, which may act in part through a mechanism involving modulation of ER homeostasis and apoptosis, particularly under conditions of cellular stresses.
Collapse
Affiliation(s)
- Evgenii Boriushkin
- Department of Ophthalmology and Biochemistry/Ross Eye Institute, University at Buffalo/SUNY, Buffalo, New York, United States SUNY Eye Institute, State University of New York, Buffalo, New York, United States
| | - Joshua J Wang
- Department of Ophthalmology and Biochemistry/Ross Eye Institute, University at Buffalo/SUNY, Buffalo, New York, United States
| | - Junhua Li
- Department of Ophthalmology and Biochemistry/Ross Eye Institute, University at Buffalo/SUNY, Buffalo, New York, United States SUNY Eye Institute, State University of New York, Buffalo, New York, United States
| | - Guangjun Jing
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Gail M Seigel
- SUNY Eye Institute, State University of New York, Buffalo, New York, United States Center for Hearing & Deafness, University at Buffalo, Buffalo/SUNY, New York, United States
| | - Sarah X Zhang
- Department of Ophthalmology and Biochemistry/Ross Eye Institute, University at Buffalo/SUNY, Buffalo, New York, United States
| |
Collapse
|
37
|
Gulyaeva NV. Brain ischemia, endoplasmic reticulum stress, and astroglial activation: new insights. J Neurochem 2015; 132:263-5. [PMID: 25586383 DOI: 10.1111/jnc.13016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
38
|
Liu H, Wang Z, Nowicki MJ. Caspase-12 mediates carbon tetrachloride-induced hepatocyte apoptosis in mice. World J Gastroenterol 2014; 20:18189-18198. [PMID: 25561786 PMCID: PMC4277956 DOI: 10.3748/wjg.v20.i48.18189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/03/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of caspase-12 and its downstream targets in carbon tetrachloride (CCl4)-induced hepatocyte apoptosis.
METHODS: The role of caspase-12 was determined by using caspase-12 knock-out (-/-) mice. CCl4 (300 μL/kg body weight) or vehicle (corn oil) was administered to caspase-12+/+ or caspase-12-/- mice as a single intraperitoneal injection. The animals were sacrificed 24 h after the CCl4 treatment. Blood was collected to evaluate liver function by the measurement of the activity of alanine aminotransferase. Liver samples were used for the measurements of reactive oxygen species using plasma malondialdehyde as biomarker, hepatocyte apoptosis was evaluated via terminal transferase-mediated dUTP nick-end labeling and controlled by morphologic study, and cytochrome C release and caspase activations were measured by Western blotting.
RESULTS: Administration of a low dose of CCl4 resulted in hepatocyte apoptosis and acute liver injury in wild-type mice. CCl4 also induced the generation of reactive oxygen species and induction of endoplasmic reticulum stress in the liver followed by activations of caspase-12, -9 and -3 as well as release of small amounts of cytochrome C. However, in the CCl4-treated caspase-12-/- mice, activation of caspase-9 and -3 were significantly attenuated (P < 0.05); no effect was seen in cytochrome C release. CCl4-induced apoptosis and liver damage was markedly reduced in caspase-12-/- mice compared to caspase-12+/+ mice (P < 0.05). The active form of caspase-8 was not detected in either caspase-12+/+ or caspase-12-/- mice. There was no significant different in the formation of reactive oxygen species in the livers of caspase-12+/+ and caspase-12-/- mice treated with CCl4.
CONCLUSION: Caspase-12 plays a pivotal role in CCl4-induced hepatic apoptosis through the activation of the downstream effector caspase-3 directly and/or indirectly via capase-9 activation.
Collapse
|
39
|
General anesthetic isoflurane modulates inositol 1,4,5-trisphosphate receptor calcium channel opening. Anesthesiology 2014; 121:528-37. [PMID: 24878495 DOI: 10.1097/aln.0000000000000316] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pharmacological evidence suggests that inhalational general anesthetics induce neurodegeneration in vitro and in vivo through overactivation of inositol trisphosphate receptor (InsP3R) Ca-release channels, but it is not clear whether these effects are due to direct modulation of channel activity by the anesthetics. METHODS Using single-channel patch clamp electrophysiology, the authors examined the gating of rat recombinant type 3 InsP3R (InsP3R-3) Ca-release channels in isolated nuclei (N = 3 to 15) from chicken lymphocytes modulated by isoflurane at clinically relevant concentrations in the absence and presence of physiological levels of the agonist inositol 1,4,5-trisphosphate (InsP3). The authors also examined the effects of isoflurane on InsP3R-mediated Ca release from the endoplasmic reticulum and changes in intracellular Ca concentration ([Ca]i). RESULTS Clinically relevant concentrations (approximately 1 minimal alveolar concentration) of the commonly used general anesthetic, isoflurane, activated InsP3R-3 channels with open probability similar to channels activated by 1 µM InsP3 (Po ≈ 0.2). This isoflurane modulation of InsP3R-3 Po depended biphasically on [Ca]i. Combination of isoflurane with subsaturating levels of InsP3 in patch pipettes resulted in at least two-fold augmentations of InsP3R-3 channel Po compared with InsP3 alone. These effects were not noted in the presence of saturating [InsP3]. Application of isoflurane to DT40 cells resulted in a 30% amplification of InsP3R-mediated [Ca]i oscillations, whereas InsP3-induced increase in [Ca]i and cleaved caspase-3 activity were enhanced by approximately 2.5-fold. CONCLUSION These results suggest that the InsP3R may be a direct molecular target of isoflurane and plays a role in the mechanisms of anesthetic-mediated pharmacological or neurotoxic effects.
Collapse
|
40
|
Liu B, Zhu Y, Zhou J, Wei Y, Long C, Chen M, Ling Y, Ge J, Zhuo Y. Endoplasmic reticulum stress promotes amyloid-beta peptides production in RGC-5 cells. Cell Stress Chaperones 2014; 19:827-35. [PMID: 24643796 PMCID: PMC4389842 DOI: 10.1007/s12192-014-0506-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 01/05/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been implicated in various neurodegenerative diseases, including Alzheimer's disease. We have previously observed amyloid production in the retina of the Tg2576 transgenic mouse model of Alzheimer's disease. In this study, we used tunicamycin-induced ER stress in RGC-5 cells, a cell line identical to the photoreceptor cell line 661W, to investigate the effect of ER stress on production of amyloid-beta (Abeta) peptides. We found that the mRNA level of amyloid-beta precursor protein (APP) remained stable, while the protein level of amyloid-beta precursor protein (APP) was decreased, the amyloid-beta precursor protein cleaving enzymes beta-site APP-cleaving enzyme 1 and presenilin 1 were upregulated, Abeta1-40 and Abeta1-42 production were increased, and reactive oxygen species production and apoptosis markers were elevated following induction of ER stress. The protein level of Abeta degradation enzymes, neprilysin, endothelin-converting enzyme 1, and endothelin-converting enzyme 2 remained unchanged during the prolonged ER stress, showing that the generation of Abeta did not result from reduction of proteolysis by these enzymes. Inclusion of group II caspase inhibitor, Z-DEVD-FMK, increased the ER stress mediated Abeta production, suggesting that they are generated by a caspase-independent mechanism. Our findings provided evidence of a role of ER stress in Abeta peptide overproduction and apoptotic pathway activation in RGC-5 cells.
Collapse
Affiliation(s)
- Bingqian Liu
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Yingting Zhu
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Jiayi Zhou
- />New England College of Optometry, Boston, MA 02115 USA
| | - Yantao Wei
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Chongde Long
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Mengfei Chen
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Yunlan Ling
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Jian Ge
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| | - Yehong Zhuo
- />State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060 China
| |
Collapse
|
41
|
Xu SY, Hu YF, Li WP, Wu YM, Ji Z, Wang SN, Li K, Pan SY. Intermittent hypothermia is neuroprotective in an in vitro model of ischemic stroke. Int J Biol Sci 2014; 10:873-81. [PMID: 25170301 PMCID: PMC4147221 DOI: 10.7150/ijbs.8868] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/14/2014] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To investigate whether the intermittent hypothermia (IH) protects neurons against ischemic insult and the potential molecular targets using an in vitro ischemic model of oxygen glucose deprivation (OGD). METHODS Fetal rat cortical neurons isolated from Day E18 rat embryos were subjected to 90-min OGD and hypothermia treatments during reoxygenation before examining the changes in microscopic morphology, cell viability, microtubule- associated protein 2 (MAP-2) release, intracellular pH value and calcium, reactive oxygen species (ROS) generation, mitochondrial membrane potential (△Ψm) and neuronal death using cell counting kit (CCK-8), enzyme-linked immunosorbent assay (ELISA), BCECF AM, Fluo-3 AM, DCFH-DA and dihydroethidium (DHE), JC-1 staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), respectively. RESULTS 90-min OGD induced morphologic abnormalities, cell viability decline, MAP-2 release, intracellular acidosis, calcium overload, increased ROS generation, △Ψm decrease and cell death in primary neurons, which was partially inhibited by continuous hypothermia (CH) and intermittent hypothermia (IH). Interestingly, 6-h CH was insufficient to reduce intracellular calcium overload and stabilize mitochondrial membrane potential (△Ψm), while 12-h CH was effective in reversing the above changes. All IH treatments (6×1 h, 4×1.5 h or 3×2 h) effectively attenuated intracellular free calcium overload, inhibited ROS production, stabilized mitochondrial membrane potential (△Ψm) and reduced delayed cell death in OGD-treated cells. However, only IH intervals longer than 1.5 h appeared to be effective in preventing cell viability loss and intracellular pH decline. CONCLUSION Both CH and IH were neuroprotective in an in vitro model of ischemic stroke, and in spite of shorter hypothermia duration, IH could provide a comparable neuroprotection to CH.
Collapse
Affiliation(s)
- Sui-yi Xu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; ; 2. Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen 518035, China
| | - Ya-fang Hu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei-pin Li
- 2. Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen 518035, China
| | - Yong-ming Wu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhong Ji
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sheng-nan Wang
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ke Li
- 3. Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Su-yue Pan
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
42
|
Zhang SX, Sanders E, Fliesler SJ, Wang JJ. Endoplasmic reticulum stress and the unfolded protein responses in retinal degeneration. Exp Eye Res 2014; 125:30-40. [PMID: 24792589 DOI: 10.1016/j.exer.2014.04.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 04/02/2014] [Accepted: 04/18/2014] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is the primary intracellular organelle responsible for protein and lipid biosynthesis, protein folding and trafficking, calcium homeostasis, and several other vital processes in cell physiology. Disturbance in ER function results in ER stress and subsequent activation of the unfolded protein response (UPR). The UPR up-regulates ER chaperones, reduces protein translation, and promotes clearance of cytotoxic misfolded proteins to restore ER homeostasis. If this vital process fails, the cell will be signaled to enter apoptosis, resulting in cell death. Sustained ER stress also can trigger an inflammatory response and exacerbate oxidative stress, both of which contribute synergistically to tissue damage. Studies performed over the past decade have implicated ER stress in a broad range of human diseases, including neurodegenerative diseases, cancer, diabetes, and vascular disorders. Several of these diseases also entail retinal dysfunction and degeneration caused by injury to retinal neurons and/or to the blood vessels that supply retinal cells with nutrients, trophic and homeostatic factors, oxygen, and other essential molecules, as well as serving as a conduit for removal of waste products and potentially toxic substances from the retina. Collectively, such injuries represent the leading cause of blindness world-wide in all age groups. Herein, we summarize recent progress on the study of ER stress and UPR signaling in retinal biology and discuss the molecular mechanisms and the potential clinical applications of targeting ER stress as a new therapeutic approach to prevent and treat neuronal degeneration in the retina.
Collapse
Affiliation(s)
- Sarah X Zhang
- Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA.
| | - Emily Sanders
- Department of Medicine, Endocrinology and Diabetes, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA; Research Service, Veterans Administration Western New York Healthcare System, Buffalo, NY, USA
| | - Joshua J Wang
- Departments of Ophthalmology and Biochemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA; SUNY Eye Institute, Buffalo, NY, USA
| |
Collapse
|
43
|
Scudeler EL, Santos DC. Side Effects of Neem Oil on the Midgut Endocrine Cells of the Green Lacewing Ceraeochrysa claveri (Navás) (Neuroptera: Chrysopidae). NEOTROPICAL ENTOMOLOGY 2014; 43:154-160. [PMID: 27193522 DOI: 10.1007/s13744-013-0191-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 12/09/2013] [Indexed: 06/05/2023]
Abstract
We described the ultrastructure of Ceraeochrysa claveri (Navás) midgut endocrine cells in larva, pupa, and adult, and evaluated the side effects of ingested neem oil, a botanical insecticide obtained from the seeds of the neem tree (Azadirachta indica), on these cells. During the larval period, C. claveri were fed (ad libitum) Diatraea saccharalis (F.) eggs treated with neem oil at concentrations of 0.5%, 1%, or 2%. Transmission electron microscopy showed that two subtypes of endocrine cells, namely granular and vesicular, occurred in the midgut epithelium during the three stages of the life cycle. Both cell types did not reach the midgut lumen and were positioned basally in the epithelium. The endocrine cells did not show extensive infoldings of the basal plasma membrane, and there were numerous secretory granules in the basal region of the cytoplasm. In the granular endocrine cells, the granules were completely filled with a dense matrix. In the vesicular endocrine cells, the main secretory products consisted of haloed vesicles. Ultrastructural examination indicated that only the granular endocrine cells exhibited signs of morphologic changes of cell injury present in all life cycle stages after the larvae were chronically exposed to neem oil by ingestion. The major cellular damage consisted of dilatation and vesiculation of the rough endoplasmic reticulum and the development of smooth endoplasmic reticulum and mitochondrial swelling. Our data suggest that cytotoxic effects on midgut endocrine cells can contribute to a generalized disruption of the physiological processes in this organ due to a general alteration of endocrine function.
Collapse
Affiliation(s)
- E L Scudeler
- Lab de Insetos, Depto de Morfologia, Instituto de Biociências, UNESP, Botucatu, SP, 18618-970, Brasil.
| | - D C Santos
- Lab de Insetos, Depto de Morfologia, Instituto de Biociências, UNESP, Botucatu, SP, 18618-970, Brasil
- Centro de Microscopia Eletrônica, Instituto de Biociências, UNESP, Botucatu, SP, Brasil
| |
Collapse
|
44
|
Li MH, Tang JP, Zhang P, Li X, Wang CY, Wei HJ, Yang XF, Zou W, Tang XQ. Disturbance of endogenous hydrogen sulfide generation and endoplasmic reticulum stress in hippocampus are involved in homocysteine-induced defect in learning and memory of rats. Behav Brain Res 2014; 262:35-41. [PMID: 24423987 DOI: 10.1016/j.bbr.2014.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 01/02/2014] [Accepted: 01/05/2014] [Indexed: 11/18/2022]
Abstract
Homocysteine (Hcy) is a risk factor for Alzheimer's disease (AD). Hydrogen sulfide (H2S) acts as an endogenous neuromodulator and neuroprotectant. It has been shown that endoplasmic reticulum (ER) stress is involved in the pathological mechanisms of the learning and memory dysfunctions and that H2S exerts its neuroprotective role via suppressing ER stress. In the present work, we explored the effects of intracerebroventricular injection of Hcy on the formation of learning and memory, the generation of endogenous H2S, and the expression of ER stress in the hippocampus of rats. We found that intracerebroventricular injection of Hcy in rats leads to learning and memory dysfunctions in the Morris water maze and novel of object recognition test and decreases in the expression of cystathionine-β-synthase, the major enzyme responsible for endogenous H2S generation, and the generation of endogenous H2S in the hippocampus of rats. We also showed that exposure of Hcy could up-regulate the expressions of glucose-regulated protein 78 (GRP78), CHOP, and cleaved caspase-12, which are the major mark proteins of ER stress, in the hippocampus of rats. Taken together, these results suggest that the disturbance of hippocampal endogenous H2S generation and the increase in ER stress in the hippocampus are related to Hcy-induced defect in learning and memory.
Collapse
Affiliation(s)
- Man-Hong Li
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China; Institute of Neuroscience, Medical College, University of South China, Hengyang, 421001 Hunan, PR China
| | - Ji-Ping Tang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China; Institute of Neuroscience, Medical College, University of South China, Hengyang, 421001 Hunan, PR China
| | - Ping Zhang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China
| | - Xiang Li
- Department of Anesthesiology, the First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China
| | - Chun-Yan Wang
- Department of Pathophysiology, Medical College, University of South China, Hengyang, 421001 Hunan, PR China
| | - Hai-Jun Wei
- Institute of Neuroscience, Medical College, University of South China, Hengyang, 421001 Hunan, PR China
| | - Xue-Feng Yang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China
| | - Wei Zou
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China.
| | - Xiao-Qing Tang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, PR China; Institute of Neuroscience, Medical College, University of South China, Hengyang, 421001 Hunan, PR China.
| |
Collapse
|
45
|
Abstract
Similar to other organisms, necrotic cell death in the nematode Caenorhabditis elegans is manifested as the catastrophic collapse of cellular homeostasis, in response to overwhelming stress that is inflicted either in the form of extreme environmental stimuli or by intrinsic insults such as the expression of proteins carrying deleterious mutations. Remarkably, necrotic cell death in C. elegans and pathological cell death in humans share multiple fundamental features and mechanistic aspects. Therefore, mechanisms mediating necrosis are also conserved across the evolutionary spectrum and render the worm a versatile tool, with the capacity to facilitate studies of human pathologies. Here, we overview necrotic paradigms that have been characterized in the nematode and outline the cellular and molecular mechanisms that mediate this mode of cell demise. In addition, we discuss experimental approaches that utilize C. elegans to elucidate the molecular underpinnings of devastating human disorders that entail necrosis.
Collapse
Affiliation(s)
- Vassiliki Nikoletopoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.
| |
Collapse
|
46
|
Rivabene R, Visentin S, Piscopo P, De Nuccio C, Crestini A, Svetoni F, Rosa P, Confaloni A. Thapsigargin affects presenilin-2 but not presenilin-1 regulation in SK-N-BE cells. Exp Biol Med (Maywood) 2013; 239:213-24. [PMID: 24363250 DOI: 10.1177/1535370213514317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Presenilin-1 (PS1) and presenilin-2 (PS2) are transmembrane proteins widely expressed in the central nervous system, which function as the catalytic subunits of γ-secretase, the enzyme that releases amyloid-β protein (Aβ) from ectodomain cleaved amyloid precursor protein (APP) by intramembrane proteolysis. Mutations in PS1, PS2, and Aβ protein precursor are involved in the etiology of familial Alzheimer's disease (FAD), while the cause of the sporadic form of AD (SAD) is still not known. However, since similar neuropathological changes have been observed in both FAD and SAD, a common pathway in the etiology of the disease has been suggested. Given that age-related deranged Ca(2+) regulation has been hypothesized to play a role in SAD pathogenesis via PS gene regulation and γ-secretase activity, we studied the in vitro regulation of PS1 and PS2 in the human neuron-like SK-N-BE cell line treated with the specific endoplasmic reticulum (ER) calcium ATPase inhibitor Thapsigargin (THG), to introduce intracellular Ca(2+) perturbations and mimic the altered Ca(2+) homeostasis observed in AD. Our results showed a consistent and significant down-regulation of PS2, while PS1 appeared to be unmodulated. These events were accompanied by oxidative stress and a number of morphological alterations suggestive of the induction of apoptotic machinery. The administration of the antioxidant N-acetylcysteine (NAC) did not revert the THG-induced effects reported, while treatment with the Ca(2+)-independent ER stressor Brefeldin A did not modulate basal PS1 and PS2 expression. Collectively, these results suggest that Ca(2+) fluctuation rather than ER stress and/or oxidative imbalance seems to play an essential role in PS2 regulation and confirm that, despite their strong homology, PS1 and PS2 could play different roles in AD.
Collapse
Affiliation(s)
- Roberto Rivabene
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299 00161 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu R, Yang G, Nonaka T, Arai T, Jia W, Cynader MS. Reducing TDP-43 aggregation does not prevent its cytotoxicity. Acta Neuropathol Commun 2013; 1:49. [PMID: 24252504 PMCID: PMC3893365 DOI: 10.1186/2051-5960-1-49] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 01/15/2023] Open
Abstract
Background TAR DNA-binding protein 43 (TDP-43) is a protein that is involved in the pathology of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Lobar Degeneration (FTLD). In patients with these neurodegenerative diseases, TDP-43 does not remain in its normal nuclear location, but instead forms insoluble aggregates in both the nucleus and cytoplasm of affected neurons. Results We used high density peptide array analysis to identify regions in TDP-43 that are bound by TDP-43 itself and designed candidate peptides that might be able to reduce TDP-43 aggregation. We found that two of the synthetic peptides identified with this approach could effectively inhibit the formation of TDP-43 protein aggregates in a concentration-dependent manner in HeLa cells in which a mutated human TDP-43 gene was overexpressed. However, despite reducing aggregation, these peptides did not reduce or prevent cell death. Similar results were observed in HeLa cells treated with arsenite. Again we found reduced aggregation, in this case of wild type TDP-43, but no difference in cell death. Conclusions Our results suggest that TDP-43 aggregation is associated with the cell death process rather than being a direct cause.
Collapse
|
48
|
Sakhrani NM, Padh H. Organelle targeting: third level of drug targeting. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:585-99. [PMID: 23898223 PMCID: PMC3718765 DOI: 10.2147/dddt.s45614] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug discovery and drug delivery are two main aspects for treatment of a variety of disorders. However, the real bottleneck associated with systemic drug administration is the lack of target-specific affinity toward a pathological site, resulting in systemic toxicity and innumerable other side effects as well as higher dosage requirement for efficacy. An attractive strategy to increase the therapeutic index of a drug is to specifically deliver the therapeutic molecule in its active form, not only into target tissue, nor even to target cells, but more importantly, into the targeted organelle, ie, to its intracellular therapeutic active site. This would ensure improved efficacy and minimize toxicity. Cancer chemotherapy today faces the major challenge of delivering chemotherapeutic drugs exclusively to tumor cells, while sparing normal proliferating cells. Nanoparticles play a crucial role by acting as a vehicle for delivery of drugs to target sites inside tumor cells. In this review, we spotlight active and passive targeting, followed by discussion of the importance of targeting to specific cell organelles and the potential role of cell-penetrating peptides. Finally, the discussion will address the strategies for drug/DNA targeting to lysosomes, mitochondria, nuclei and Golgi/endoplasmic reticulum.
Collapse
Affiliation(s)
- Niraj M Sakhrani
- Department of Cell and Molecular Biology, BV Patel Pharmaceutical Education and Research Development (PERD) Centre, Gujarat, India
| | | |
Collapse
|
49
|
Avila MF, Cabezas R, Torrente D, Gonzalez J, Morales L, Alvarez L, Capani F, Barreto GE. Novel interactions of GRP78: UPR and estrogen responses in the brain. Cell Biol Int 2013; 37:521-32. [DOI: 10.1002/cbin.10058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/22/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Marco Fidel Avila
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Ricardo Cabezas
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Daniel Torrente
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Janneth Gonzalez
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Ludis Morales
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Lisandro Alvarez
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), Facultad de Medicina, UBA-CONICET; Marcelo T. de Alvear 2270, C1122AAJ Buenos Aires; Argentina
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), Facultad de Medicina, UBA-CONICET; Marcelo T. de Alvear 2270, C1122AAJ Buenos Aires; Argentina
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| |
Collapse
|
50
|
Saleh A, Schapansky J, Smith DR, Young N, Odero GL, Aulston B, Fernyhough P, Glazner GW. Normalization of NF-κB activity in dorsal root ganglia neurons cultured from diabetic rats reverses neuropathy-linked markers of cellular pathology. Exp Neurol 2012; 241:169-78. [PMID: 23159890 DOI: 10.1016/j.expneurol.2012.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 10/05/2012] [Accepted: 11/06/2012] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS Dorsal root ganglia (DRG) sensory neurons cultured from 3 to 5 month streptozotocin (STZ)-induced diabetic rats exhibit structural and biochemical changes seen in peripheral nerve fibers in vivo, including axonal swellings, oxidative damage, reduced axonal sprouting, and decreased NF-κB activity. NF-κB is a transcription factor required by DRG neurons for survival and plasticity, and regulates transcription of antioxidant proteins (e.g. MnSOD). We hypothesized that the diabetes-induced decrease in NF-κB activity in DRG contributes to pathological phenomena observed in cultured DRG neurons from diabetic rats. METHODS NF-κB localization was assessed in intact DRG and neuron cultures using immunostaining. NF-κB activity was manipulated in sensory neuron cultures derived from age-matched normal or 3-5 month STZ-diabetic rats using pharmacological means and lentiviral expression of shRNA. The impact of diabetes and altered NF-κB activity on neuronal phenotype involved analysis of neurite outgrowth, neurite morphology, oxidative stress (lipid peroxidation) and expression of MnSOD. RESULTS STZ-induced diabetes caused a significant decrease in nuclear localization of NF-κB subunits p50 and c-rel, but no change in p65 in intact DRG. Inhibition of NF-κB in normal neuron cultures significantly increased axonal swellings and oxidative stress, and reduced both neurite outgrowth and expression of MnSOD. These phenomena mimicked markers of pathology in cultured DRG neurons from diabetic rats. Enhancement of NF-κB activity in cultured diabetic DRG neurons ameliorated the sub-optimal neurite outgrowth and MnSOD levels triggered by diabetes. Exogenous insulin enhanced nuclear localization of p50 and c-rel but not p65 in diabetic neuronal cultures. CONCLUSION/INTERPRETATION The diabetes-induced decrease of nuclear localization of NF-κB subunits p50 and c-rel in DRG contributes to development of in vitro markers of peripheral neuropathy, possibly through impaired mitochondrial ROS scavenging by deficient MnSOD.
Collapse
Affiliation(s)
- A Saleh
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | |
Collapse
|