51
|
Ahmed M, Machado PM, Miller A, Spicer C, Herbelin L, He J, Noel J, Wang Y, McVey AL, Pasnoor M, Gallagher P, Statland J, Lu CH, Kalmar B, Brady S, Sethi H, Samandouras G, Parton M, Holton JL, Weston A, Collinson L, Taylor JP, Schiavo G, Hanna MG, Barohn RJ, Dimachkie MM, Greensmith L. Targeting protein homeostasis in sporadic inclusion body myositis. Sci Transl Med 2016; 8:331ra41. [PMID: 27009270 PMCID: PMC5043094 DOI: 10.1126/scitranslmed.aad4583] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/04/2016] [Indexed: 11/02/2022]
Abstract
Sporadic inclusion body myositis (sIBM) is the commonest severe myopathy in patients more than 50 years of age. Previous therapeutic trials have targeted the inflammatory features of sIBM but all have failed. Because protein dyshomeostasis may also play a role in sIBM, we tested the effects of targeting this feature of the disease. Using rat myoblast cultures, we found that up-regulation of the heat shock response with arimoclomol reduced key pathological markers of sIBM in vitro. Furthermore, in mutant valosin-containing protein (VCP) mice, which develop an inclusion body myopathy, treatment with arimoclomol ameliorated disease pathology and improved muscle function. We therefore evaluated arimoclomol in an investigator-led, randomized, double-blind, placebo-controlled, proof-of-concept trial in sIBM patients and showed that arimoclomol was safe and well tolerated. Although arimoclomol improved some IBM-like pathology in the mutant VCP mouse, we did not see statistically significant evidence of efficacy in the proof-of-concept patient trial.
Collapse
Affiliation(s)
- Mhoriam Ahmed
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Pedro M Machado
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Adrian Miller
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Charlotte Spicer
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Laura Herbelin
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - Jianghua He
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Janelle Noel
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yunxia Wang
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - April L McVey
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - Mamatha Pasnoor
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - Philip Gallagher
- Department of Health, Sport, and Exercise Science, The University of Kansas, Lawrence, KS 66045-7567, USA
| | - Jeffrey Statland
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - Ching-Hua Lu
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Bernadett Kalmar
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Stefen Brady
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Huma Sethi
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, UCL Hospitals, Queen Square, London WC1N 3BG, UK
| | - George Samandouras
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, UCL Hospitals, Queen Square, London WC1N 3BG, UK
| | - Matt Parton
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Janice L Holton
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Anne Weston
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Lucy Collinson
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - J Paul Taylor
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Giampietro Schiavo
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Michael G Hanna
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Richard J Barohn
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA
| | - Mazen M Dimachkie
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Mail Stop 2012, Kansas City, KS 66160, USA.
| | - Linda Greensmith
- Medical Research Council (MRC) Centre for Neuromuscular Diseases, University College London (UCL) Institute of Neurology, Queen Square, London WC1N 3BG, UK. Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
52
|
Stress Response Leading to Resistance in Glioblastoma-The Need for Innovative Radiotherapy (iRT) Concepts. Cancers (Basel) 2016; 8:cancers8010015. [PMID: 26771644 PMCID: PMC4728462 DOI: 10.3390/cancers8010015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/23/2015] [Accepted: 01/08/2016] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is the most common and most aggressive malignant primary brain tumor in adults. In spite of multimodal therapy concepts, consisting of surgery, radiotherapy and chemotherapy, the median survival, merely 15–18 months, is still poor. Mechanisms for resistance of GBM to radio(chemo)therapy are not fully understood yet and due to the genetic heterogeneity within the tumor including radiation-resistant tumor stem cells, there are several factors leading to therapy failure. Recent research revealed that, hypoxia during radiation and miRNAs may adversely affect the therapeutic response to radiotherapy. Further molecular alterations and prognostic markers like the DNA-repair protein O6-methylguanine-DNA methyltransferase (MGMT), anti-apoptotic molecular chaperones, and/or the activity of aldehyde dehydrogenase 1 (ALDH1) have also been identified to play a role in the sensitivity to cytostatic agents. Latest approaches in the field of radiotherapy to use particle irradiation or dose escalation strategies including modern molecular imaging, however, need further evaluation with regard to long-term outcome. In this review we focus on current information about the mechanisms and markers that mediate resistance to radio(chemo)therapy, and discuss the opportunities of Innovative Radiotherapy (iRT) concepts to improve treatment options for GBM patients.
Collapse
|
53
|
Abstract
All living organisms sense and respond to harmful changes in their intracellular and extracellular environment through complex signaling pathways that lead to changes in gene expression and cellular function in order to maintain homeostasis. Long non-coding RNAs (lncRNAs), a large and heterogeneous group of functional RNAs, play important roles in cellular response to stressful conditions. lncRNAs constitute a significant fraction of the genes differentially expressed in response to diverse stressful stimuli and, once induced, contribute to the regulation of downstream cellular processes, including feedback regulation of key stress response proteins. While many lncRNAs seem to be induced in response to a specific stress, there is significant overlap between lncRNAs induced in response to different stressful stimuli. In addition to stress-induced RNAs, several constitutively expressed lncRNAs also exert a strong regulatory impact on the stress response. Although our understanding of the contribution of lncRNAs to the cellular stress response is still highly rudimentary, the existing data point to the presence of a complex network of lncRNAs, miRNAs, and proteins in regulation of the cellular response to stress.
Collapse
Affiliation(s)
- Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Alberto Valencia-Hipólito
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| |
Collapse
|
54
|
McCallister C, Kdeiss B, Nikolaidis N. Biochemical characterization of the interaction between HspA1A and phospholipids. Cell Stress Chaperones 2016; 21:41-53. [PMID: 26342809 PMCID: PMC4679732 DOI: 10.1007/s12192-015-0636-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/25/2015] [Accepted: 08/31/2015] [Indexed: 01/15/2023] Open
Abstract
Seventy-kilodalton heat shock proteins (Hsp70s) are molecular chaperones essential for maintaining cellular homeostasis. Apart from their indispensable roles in protein homeostasis, specific Hsp70s localize at the plasma membrane and bind to specific lipids. The interaction of Hsp70s with lipids has direct physiological outcomes including lysosomal rescue, microautophagy, and promotion of cell apoptosis. Despite these essential functions, the Hsp70-lipid interactions remain largely uncharacterized. In this study, we characterized the interaction of HspA1A, an inducible Hsp70, with five phospholipids. We first used high concentrations of potassium and established that HspA1A embeds in membranes when bound to all anionic lipids tested. Furthermore, we found that protein insertion is enhanced by increasing the saturation level of the lipids. Next, we determined that the nucleotide-binding domain (NBD) of the protein binds to lipids quantitatively more than the substrate-binding domain (SBD). However, for all lipids tested, the full-length protein is necessary for embedding. We also used calcium and reaction buffers equilibrated at different pH values and determined that electrostatic interactions alone may not fully explain the association of HspA1A with lipids. We then determined that lipid binding is inhibited by nucleotide-binding, but it is unaffected by protein-substrate binding. These results suggest that the HspA1A lipid-association is specific, depends on the physicochemical properties of the lipid, and is mediated by multiple molecular forces. These mechanistic details of the Hsp70-lipid interactions establish a framework of possible physiological functions as they relate to chaperone regulation and localization.
Collapse
Affiliation(s)
- Chelsea McCallister
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Brianna Kdeiss
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA.
| |
Collapse
|
55
|
Tsai LK, Chen CL, Tsai YC, Ting CH, Chien YH, Lee NC, Hwu WL. Hypothermia improves disease manifestations in SMA mice via SMN augmentation. Hum Mol Genet 2015; 25:631-41. [PMID: 26647309 DOI: 10.1093/hmg/ddv500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/01/2015] [Indexed: 11/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a progressive motor neuron disease caused by a deficiency of survival motor neuron (SMN) protein. In this study, we evaluated the efficacy of intermittent transient hypothermia in a mouse model of SMA. SMA mice were exposed to ice for 50 s to achieve transient hypothermia (below 25°C) daily beginning on postnatal day 1. Neonatal SMA mice (Smn(-/-)SMN2(+/-)) who received daily transient hypothermia exhibited reduced motor neuron degeneration and muscle atrophy and preserved the architecture of neuromuscular junction when compared with untreated controls at day 8 post-treatment. Daily hypothermia also prolonged the lifespan, increased body weight and improved motor coordination in SMA mice. Quantitative polymerase chain reaction and western blot analyses showed that transient hypothermia led to an increase in SMN transcript and protein levels in the spinal cord and brain. In in vitro studies using an SMN knockdown motor neuron-like cell-line, transient hypothermia increased intracellular SMN protein expression and length of neurites, confirming the direct effect of hypothermia on motor neurons. These data indicate that the efficacy of intermittent transient hypothermia in improving outcome in an SMA mouse model may be mediated, in part, via an upregulation of SMN levels in the motor neurons.
Collapse
Affiliation(s)
- Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan,
| | - Chien-Lin Chen
- Department of Neurology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yi-Chieh Tsai
- Department of Neurology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Chen-Hung Ting
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan and
| | - Yin-Hsio Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Ni-Chong Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
56
|
Shabbir A, Bianchetti E, Cargonja R, Petrovic A, Mladinic M, Pilipović K, Nistri A. Role of HSP70 in motoneuron survival after excitotoxic stress in a rat spinal cord injury modelin vitro. Eur J Neurosci 2015; 42:3054-65. [DOI: 10.1111/ejn.13108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Ayisha Shabbir
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Elena Bianchetti
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Renato Cargonja
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Antonela Petrovic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Miranda Mladinic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Kristina Pilipović
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Andrea Nistri
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| |
Collapse
|
57
|
Dattilo S, Mancuso C, Koverech G, Di Mauro P, Ontario ML, Petralia CC, Petralia A, Maiolino L, Serra A, Calabrese EJ, Calabrese V. Heat shock proteins and hormesis in the diagnosis and treatment of neurodegenerative diseases. Immun Ageing 2015; 12:20. [PMID: 26543490 PMCID: PMC4634585 DOI: 10.1186/s12979-015-0046-8] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
Modulation of endogenous cellular defense mechanisms via the vitagene system represents an innovative approach to therapeutic intervention in diseases causing chronic tissue damage, such as in neurodegeneration. The possibility of high-throughoutput screening using proteomic techniques, particularly redox proteomics, provide more comprehensive overview of the interaction of proteins, as well as the interplay among processes involved in neuroprotection. Here by introducing the hormetic dose response concept, the mechanistic foundations and applications to the field of neuroprotection, we discuss the emerging role of heat shock protein as prominent member of vitagene network in neuroprotection and redox proteomics as a tool for investigating redox modulation of stress responsive vitagenes. Hormetic mechanisms are reviewed as possibility of targeted therapeutic manipulation in a cell-, tissue- and/or pathway-specific manner at appropriate points in the neurodegenerative disease process.
Collapse
Affiliation(s)
- Sandro Dattilo
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | - Cesare Mancuso
- />Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Guido Koverech
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | - Paola Di Mauro
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Maria Laura Ontario
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | | | - Antonino Petralia
- />Department of Clinical and Experimental Medicine, School of Medicine, University of Catania, Catania, Italy
| | - Luigi Maiolino
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Agostino Serra
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Edward J. Calabrese
- />Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, MA USA
| | - Vittorio Calabrese
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| |
Collapse
|
58
|
Eukaryotic aggresomes: from a model of conformational diseases to an emerging type of immobilized biocatalyzers. Appl Microbiol Biotechnol 2015; 100:559-69. [DOI: 10.1007/s00253-015-7107-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 12/28/2022]
|
59
|
HspA1A, a 70-kDa heat shock protein, differentially interacts with anionic lipids. Biochem Biophys Res Commun 2015; 467:835-40. [PMID: 26476215 DOI: 10.1016/j.bbrc.2015.10.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/10/2015] [Indexed: 12/22/2022]
Abstract
HspA1A, a 70-kDa heat shock protein, binds to specific lipids. This interaction allows HspA1A to associate with the plasma and other cellular membranes, where it regulates many vital functions like immunity, membrane stabilization, autophagy, and apoptosis. However, the molecular mechanism of the HspA1A-lipid interactions has yet to be fully characterized. Therefore, in this study, we characterized the interaction of HspA1A with three lipids, bis-(monoacylglycero)-phosphate, cardiolipin, and sulfatide. Our results revealed that, first, HspA1A embeds in membranes when bound to liposomes composed of cardiolipin and sulfatide. Second, the binding of HspA1A to lipids is complex and although important, electrostatic interactions alone cannot fully explain the observed binding. Third, the two HspA1A domains, the nucleotide-binding domain and the substrate-binding domain, differentially bind to lipids in a lipid-specific manner. Fourth, HspA1A lipid-binding is reduced by the presence of nucleotides, but it is unaffected by the presence of a peptide-substrate. These observations suggest that HspA1A binds to lipids via a multi-step mechanism and this interaction depends on the specific physicochemical properties of the lipid. We speculate that the association of HspA1A with lipids like the mitochondrial cardiolipin, which is an organelle marker, may facilitate the translocation and localized function of the molecular chaperone to particular sub-cellular compartments.
Collapse
|
60
|
Xia C, Cai Y, Lin Y, Guan R, Xiao G, Yang J. MiR-133b-5p regulates the expression of the heat shock protein 70 during rat neuronal cell apoptosis induced by the gp120 V3 loop peptide. J Med Virol 2015; 88:437-47. [PMID: 26280272 DOI: 10.1002/jmv.24355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2015] [Indexed: 12/28/2022]
Abstract
Neuronal cell dysfunction and apoptosis, the main causes of HIV-associated dementia, and its underlying mechanism are important unsolved health problems. Many research reports suggest that miRNAs regulate HIV-1-induced apoptosis. We used the HIV-1 gp120 V3 Loop peptide to induce primary rat cortical neurons apoptosis. Next, we used a microRNA microarray to identify the significant changes of miRNA in the rat cortical neurons treated with the gp120 V3 loop peptide. We used western blot and real-time PCR to measure the regulation of heat shock protein 70 by rno-miR-133b-5p. In response to the gp120 V3 loop peptide treatment, rat cortical neurons exhibited 11 up-regulated and 21 down-regulated miRNAs. We further examined miR-133b-5p, a microRNA that was up-regulated more than 118-fold. In addition, both HSP70 mRNA and protein expression were dose-dependent in rats cortical neurons treated with gp120 V3 loop peptide for 48 hr. MiR-133b-5p could regulate heat shock protein 70 (HSP70) at both transcription and translation levels. Rno-miR-133b-5p might be less significant for the gp120 V3 loop peptide induced neuron apoptosis. Thus, we discovered a potential new target for the regulation of HIV-1 gp120- induced apoptosis.
Collapse
Affiliation(s)
- Chenglai Xia
- Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P.R. China
| | - Yantao Cai
- Department of Dermatology and Rheumatology, Foshan Maternity & Child Heath Care Hospital, Foshan, 528000, China
| | - Yuyi Lin
- Department of Reproductive Medicine Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P.R. China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou Medical University at Guangzhou, 63 Duobao Road, Guangzhou, GD, 510150, China
| | - Ronghua Guan
- Department of Reproductive Medicine Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P.R. China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou Medical University at Guangzhou, 63 Duobao Road, Guangzhou, GD, 510150, China
| | - Guohong Xiao
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou Medical University at Guangzhou, 63 Duobao Road, Guangzhou, GD, 510150, China
| | - Jie Yang
- Department of Reproductive Medicine Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, P.R. China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou Medical University at Guangzhou, 63 Duobao Road, Guangzhou, GD, 510150, China
| |
Collapse
|
61
|
Yacila G, Sari Y. Potential therapeutic drugs and methods for the treatment of amyotrophic lateral sclerosis. Curr Med Chem 2015; 21:3583-93. [PMID: 24934355 DOI: 10.2174/0929867321666140601162710] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 04/08/2014] [Accepted: 05/26/2014] [Indexed: 12/13/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder caused by damage of motoneurons leading to paralysis state and long term disability. Riluzole is currently the only FDA-approved drug for the treatment of ALS. The proposed mechanisms of ALS include glutamate excitotoxicity, oxidative stress, mitochondrial dysfunction, protein aggregation, SOD1 accumulations, and neuronal death. In this review, we discuss potential biomarkers for the identification of patients with ALS. We further emphasize potential therapy involving the uses of neurotrophic factors such as IGFI, GDNF, VEGF, ADNF-9, colivelin and angiogenin in the treatment of ALS. Moreover, we described several existing drugs such as talampanel, ceftriaxone, pramipexole, dexpramipexole and arimoclomol potential compounds for the treatment of ALS. Interestingly, the uses of stem cell therapy and immunotherapy are promising for the treatment of ALS.
Collapse
Affiliation(s)
| | - Y Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Health Science Campus, 3000 Arlington Avenue, Toledo, OH 43614. USA.
| |
Collapse
|
62
|
Fröhlich D, Kuo WP, Frühbeis C, Sun JJ, Zehendner CM, Luhmann HJ, Pinto S, Toedling J, Trotter J, Krämer-Albers EM. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0510. [PMID: 25135971 DOI: 10.1098/rstb.2013.0510] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Exosomes are small membranous vesicles of endocytic origin that are released by almost every cell type. They exert versatile functions in intercellular communication important for many physiological and pathological processes. Recently, exosomes attracted interest with regard to their role in cell-cell communication in the nervous system. We have shown that exosomes released from oligodendrocytes upon stimulation with the neurotransmitter glutamate are internalized by neurons and enhance the neuronal stress tolerance. Here, we demonstrate that oligodendroglial exosomes also promote neuronal survival during oxygen-glucose deprivation, a model of cerebral ischaemia. We show the transfer from oligodendrocytes to neurons of superoxide dismutase and catalase, enzymes which are known to help cells to resist oxidative stress. Additionally, we identify various effects of oligodendroglial exosomes on neuronal physiology. Electrophysiological analysis using in vitro multi-electrode arrays revealed an increased firing rate of neurons exposed to oligodendroglial exosomes. Moreover, gene expression analysis and phosphorylation arrays uncovered differentially expressed genes and altered signal transduction pathways in neurons after exosome treatment. Our study thus provides new insight into the broad spectrum of action of oligodendroglial exosomes and their effects on neuronal physiology. The exchange of extracellular vesicles between neural cells may exhibit remarkable potential to impact brain performance.
Collapse
Affiliation(s)
- Dominik Fröhlich
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Wen Ping Kuo
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Carsten Frühbeis
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Jyh-Jang Sun
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany Neuro-Electronics Research Flanders, 3001 Leuven, Belgium
| | - Christoph M Zehendner
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany ZIM III, Department of Cardiology, Goethe University Frankfurt, 60389 Frankfurt, Germany
| | - Heiko J Luhmann
- Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany Institute of Physiology, University Medical Center, 55128 Mainz, Germany
| | - Sheena Pinto
- Division of Developmental Immunology, DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Joern Toedling
- Institute of Molecular Biology gGmbH, 55128 Mainz, Germany
| | - Jacqueline Trotter
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Eva-Maria Krämer-Albers
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| |
Collapse
|
63
|
Son SJ, Lee KS, Chung JH, Chang KJ, Roh HW, Kim SH, Jin T, Back JH, Kim HJ, Lee Y, Choi SH, Noh JS, Lim KY, Chung YK, Hong CH, Oh BH. Increased plasma levels of heat shock protein 70 associated with subsequent clinical conversion to mild cognitive impairment in cognitively healthy elderly. PLoS One 2015; 10:e0119180. [PMID: 25768018 PMCID: PMC4359110 DOI: 10.1371/journal.pone.0119180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/11/2015] [Indexed: 11/24/2022] Open
Abstract
Background and Aims Heat shock proteins (HSPs) have been regarded as cytoprotectants that protect brain cells during the progression of neurodegenerative diseases and from damage resulting from cerebral ischemia. In this study, we assessed the association between plasma HSP 70/27 levels and cognitive decline. Methods Among participants in the community-based cohort study of dementia called the Gwangju Dementia and Mild Cognitive Impairment Study, subjects without cognitive impairment at baseline, who then either remained without impairment (non-conversion group), or suffered mild cognitive impairment (MCI) (conversion group) (non-conversion group, N = 36; conversion group, N = 30) were analyzed. Results After a five to six year follow-up period, comparison of the plasma HSP 70 and HSP 27 levels of the two groups revealed that only the plasma HSP 70 level was associated with a conversion to MCI after adjustments for age, gender, years of education, follow-up duration, APOE e4, hypertension, and diabetes (repeated measure analysis of variance: F = 7.59, p = 0.008). Furthermore, an increase in plasma HSP 70 level was associated with cognitive decline in language and executive function (linear mixed model: Korean Boston Naming Test, -0.426 [-0.781, -0.071], p = 0.019; Controlled Oral Word Association Test, -0.176 [-0.328, -0.023], p = 0.024; Stroop Test, -0.304 [-0.458, -0.150], p<0.001). Conclusions These findings suggest that the plasma HSP 70 level may be related to cognitive decline in the elderly.
Collapse
Affiliation(s)
- Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Kang Soo Lee
- Department of Psychiatry, CHA Gangnam Medical Center, CHA University, Nonhyon-ro, Gangnam-gu, Seoul 135–081, Republic of Korea
| | - Ji Hyung Chung
- Department of Applied Bioscience, College of Life Science, CHA University, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Republic of Korea
| | - Ki Jung Chang
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Soo Hyun Kim
- Graduate Program in Science for Aging, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120–752, Republic of Korea
| | - Taewon Jin
- Graduate Program in Science for Aging, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120–752, Republic of Korea
| | - Joung Hwan Back
- Health Insurance Policy Research Institute, National Health Insurance Service, 311 Dongmak-ro, Mapo-gu, Seoul 121–749, Republic of Korea
| | - Hyun Jung Kim
- Department of Psychiatry, National Medical Center, 245 Eulji-ro, Jung-gu, Seoul 100–799, Republic of Korea
| | - Yunhwan Lee
- Department of Preventive Medicine and Public Health, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University College of Medicine, 27 Inhang-ro, Jung-gu, Incheon 400–711, Republic of Korea
| | - Jai Sung Noh
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Ki Young Lim
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Young Ki Chung
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 443–380, Republic of Korea
- * E-mail: (CHH); (BHO)
| | - Byoung Hoon Oh
- Department of Psychiatry and Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120–752, Republic of Korea
- * E-mail: (CHH); (BHO)
| |
Collapse
|
64
|
Karunanithi S, Brown IR. Heat shock response and homeostatic plasticity. Front Cell Neurosci 2015; 9:68. [PMID: 25814928 PMCID: PMC4357293 DOI: 10.3389/fncel.2015.00068] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/17/2015] [Indexed: 11/13/2022] Open
Abstract
Heat shock response and homeostatic plasticity are mechanisms that afford functional stability to cells in the face of stress. Each mechanism has been investigated independently, but the link between the two has not been extensively explored. We explore this link. The heat shock response enables cells to adapt to stresses such as high temperature, metabolic stress and reduced oxygen levels. This mechanism results from the production of heat shock proteins (HSPs) which maintain normal cellular functions by counteracting the misfolding of cellular proteins. Homeostatic plasticity enables neurons and their target cells to maintain their activity levels around their respective set points in the face of stress or disturbances. This mechanism results from the recruitment of adaptations at synaptic inputs, or at voltage-gated ion channels. In this perspective, we argue that heat shock triggers homeostatic plasticity through the production of HSPs. We also suggest that homeostatic plasticity is a form of neuroprotection.
Collapse
Affiliation(s)
- Shanker Karunanithi
- School of Medical Science, Griffith University QLD, Australia ; Menzies Health Institute of Queensland, Griffith University QLD, Australia
| | - Ian R Brown
- Department of Biological Sciences, Centre for the Neurobiology of Stress, University of Toronto Scarborough Toronto, ON, Canada
| |
Collapse
|
65
|
Kraynik SM, Gabanic A, Anthony SR, Kelley M, Paulding WR, Roessler A, McGuinness M, Tranter M. The stress-induced heat shock protein 70.3 expression is regulated by a dual-component mechanism involving alternative polyadenylation and HuR. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:688-96. [PMID: 25727182 DOI: 10.1016/j.bbagrm.2015.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/31/2015] [Accepted: 02/21/2015] [Indexed: 10/23/2022]
Abstract
Heat shock protein 70.3 (Hsp70.3) expression increases in response to cellular stress and plays a cytoprotective role. We have previously shown that Hsp70.3 expression is controlled through coordinated post-transcriptional regulation by miRNAs and alternative polyadenylation (APA), and APA-mediated shortening of the Hsp70.3 3'-UTR facilitates increased protein expression. A stress-induced increase in Hsp70.3 mRNA and protein expression is accompanied by alternative polyadenylation (APA)-mediated truncation of the 3'UTR of the Hsp70.3 mRNA transcript. However, the role that APA plays in stress-induced expression of Hsp70.3 remains unclear. Our results show that APA-mediated truncation of the Hsp70.3 3'UTR increases protein expression through enhanced polyribosome loading. Additionally, we demonstrate that the RNA binding protein HuR, which has been previously shown to play a role in mediating APA, is necessary for heat shock mediated increase in Hsp70.3 mRNA and protein. However, it is somewhat surprising to note that HuR does not play a role in APA of the Hsp70.3 mRNA, and these two regulatory events appear to be mutually exclusive regulators of Hsp70.3 expression. These results not only provide important insight to the regulation of stress response genes following heat shock, but also contribute an enhanced understanding of how alternative polyadenylation contributes to gene regulation.
Collapse
Affiliation(s)
- Stephen M Kraynik
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Andrew Gabanic
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Sarah R Anthony
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Melissa Kelley
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | | | - Anne Roessler
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Michael McGuinness
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Michael Tranter
- Dept. of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
66
|
Khalouei S, Chow AM, Brown IR. Localization of heat shock protein HSPA6 (HSP70B') to sites of transcription in cultured differentiated human neuronal cells following thermal stress. J Neurochem 2014; 131:743-54. [DOI: 10.1111/jnc.12970] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/05/2014] [Accepted: 10/06/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Sam Khalouei
- Centre for the Neurobiology of Stress; Department of Biological Sciences; University of Toronto Scarborough; Toronto Ontario Canada
| | - Ari M. Chow
- Centre for the Neurobiology of Stress; Department of Biological Sciences; University of Toronto Scarborough; Toronto Ontario Canada
| | - Ian R. Brown
- Centre for the Neurobiology of Stress; Department of Biological Sciences; University of Toronto Scarborough; Toronto Ontario Canada
| |
Collapse
|
67
|
Chow AM, Tang DWF, Hanif A, Brown IR. Localization of heat shock proteins in cerebral cortical cultures following induction by celastrol. Cell Stress Chaperones 2014; 19:845-51. [PMID: 24700193 PMCID: PMC4389844 DOI: 10.1007/s12192-014-0508-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 12/28/2022] Open
Abstract
Hsp70, Hsp32, and Hsp27 were induced by celastrol in rat cerebral cortical cultures at dosages that did not affect cell viability. Pronounced differences were observed in the cellular localization of these heat shock proteins in cell types of cerebral cortical cultures. Celastrol-induced Hsp70 localized to the cell body and cellular processes of neurons that were identified by neuron-specific βIII-tubulin. Hsp70 was not detected in adjacent GFAP-positive glial cells that demonstrated a strong signal for Hsp27 and Hsp32 in both glial cell bodies and cellular processes. Cells in the cerebral cortex region of the brain are selectively impacted during the progression of Alzheimer's disease which is a "protein misfolding disorder." Heat shock proteins provide a line of defense against misfolded, aggregation-prone proteins. Celastrol is a potential agent to counter this neurodegenerative disorder as recent evidence indicates that in vivo administration of celastrol in a transgenic model of Alzheimer's reduces an important neuropathological hallmark of this disease, namely, amyloid beta pathology that involves protein aggregation.
Collapse
Affiliation(s)
- Ari M. Chow
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Derek W. F. Tang
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Asad Hanif
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Ian R. Brown
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| |
Collapse
|
68
|
Malaspina A, Puentes F, Amor S. Disease origin and progression in amyotrophic lateral sclerosis: an immunology perspective. Int Immunol 2014; 27:117-29. [DOI: 10.1093/intimm/dxu099] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
69
|
Lei M, Dong D, Mu S, Pan YH, Zhang S. Comparison of brain transcriptome of the greater horseshoe bats (Rhinolophus ferrumequinum) in active and torpid episodes. PLoS One 2014; 9:e107746. [PMID: 25251558 PMCID: PMC4174523 DOI: 10.1371/journal.pone.0107746] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 08/21/2014] [Indexed: 01/03/2023] Open
Abstract
Hibernation is an energy-saving strategy which is widely adopted by heterothermic mammals to survive in the harsh environment. The greater horseshoe bat (Rhinolophus ferrumequinum) can hibernate for a long period in the hibernation season. However, the global gene expression changes between hibernation and non-hibernation season in the greater horseshoe bat remain largely unknown. We herein reported a comprehensive survey of differential gene expression in the brain between winter hibernating and summer active greater horseshoe bats using next-generation sequencing technology. A total of 90,314,174 reads were generated and we identified 1,573 differentially expressed genes between active and torpid states. Interestingly, we found that differentially expressed genes are over-represented in some GO categories (such as metabolic suppression, cellular stress responses and oxidative stress), which suggests neuroprotective strategies might play an important role in hibernation control mechanisms. Our results determined to what extent the brain tissue of the greater horseshoe bats differ in gene expression between summer active and winter hibernating states and provided comprehensive insights into the adaptive mechanisms of bat hibernation.
Collapse
Affiliation(s)
- Ming Lei
- Institute of Molecular Ecology and Evolution, SKLEC & IECR, East China Normal University, Shanghai, China
| | - Dong Dong
- Institute of Molecular Ecology and Evolution, SKLEC & IECR, East China Normal University, Shanghai, China
- * E-mail: (DD); (SZ)
| | - Shuo Mu
- Institute of Molecular Ecology and Evolution, SKLEC & IECR, East China Normal University, Shanghai, China
| | - Yi-Hsuan Pan
- Institute of Molecular Ecology and Evolution, SKLEC & IECR, East China Normal University, Shanghai, China
| | - Shuyi Zhang
- Institute of Molecular Ecology and Evolution, SKLEC & IECR, East China Normal University, Shanghai, China
- * E-mail: (DD); (SZ)
| |
Collapse
|
70
|
Heat shock proteins at the crossroads between cancer and Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:239164. [PMID: 25147790 PMCID: PMC4131517 DOI: 10.1155/2014/239164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/12/2014] [Indexed: 12/31/2022]
Abstract
Heat shock proteins 70 and heat shock proteins 90 (Hsp70/90) have been implicated in many crucial steps of carcinogenesis: stabilizing oncogenic proteins, inhibiting programmed cell death and replicative senescence, induction of tumor angiogenesis, and activation of the invasion and metastasis. Plenty of cancer related proteins have the ability of regulating the expression of Hsp70/90 through heat shock factor 1. Cancer and Alzheimer's disease (AD) have plenty of overlapping regions in molecular genetics and cell biology associated with Hsp70/90. The Hsp70, as a protein stabilizer, has a cellular protection against neurodegeneration of the central nervous system, while Hsp90 promote neurodegenerative disorders indirectly through regulating the expression of Hsp70 and other chaperones. All these make existing anticancer drugs target Hsp70/90 which might be used in AD therapy.
Collapse
|
71
|
Talla V, Porciatti V, Chiodo V, Boye SL, Hauswirth WW, Guy J. Gene therapy with mitochondrial heat shock protein 70 suppresses visual loss and optic atrophy in experimental autoimmune encephalomyelitis. Invest Ophthalmol Vis Sci 2014; 55:5214-26. [PMID: 25015358 DOI: 10.1167/iovs.14-14688] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE To rescue visual loss and optic neuropathy in experimental autoimmune encephalomyelitis (EAE). METHODS Encephalomyelitis was induced in mice that received intravitreal injections of AAV2-mtHSP70Flag or AAV2-Cox8-mCherry. Additional mice were injected with AAV2-Cox8-mCherry, but not sensitized for EAE. Visual function was assessed by pattern electroretinograms (PERG) at 1, 3, and 6 months post injection (MPI). Optical coherence tomography (OCT) evaluated the thickness of the inner plexiform layer + nerve fiber layers at 1, 3, and 6 MPI. Retinas and optic nerves (ONs) of mice euthanized 6 MPI were processed for light and electron microscopy. Expression of mtHSP70Flag in the retina and ONs was evaluated by RT-PCR, immunofluorescence, and Western blotting. The activities of respiratory complexes I and III, as well as mitochondrial protein import were quantitated. RESULTS Expression: immunofluorescence revealed punctate and perinuclear expression of mtHSP70Flag that colocalized with mitochondrial porin in thy1.2 labeled retinal ganglion cells (RGCs). Immunoblotting and RT-PCR confirmed mtHSP70Flag expression in the retina and ON. Rescue: treatment with mtHSP70Flag resulted in a 44% increase in PERG amplitude and less delays in latency relative to the EAE-mCherry group that also showed progressive inner retinal thinning. At 6 MPI, the almost 50% loss of RGCs and optic nerve axons in EAE mice was suppressed by mtHSP70Flag. In addition, retinas of EAE-mtHSP70Flag mice showed nearly complete rescue of complex I and III activities that was reduced by one-third in the EAE-mCherry retinas. Lastly, reductions in import of COX8-mCherry into mitochondria of mice sensitized for EAE improved by 30% with mtHSP70Flag gene therapy. CONCLUSIONS Mitochondrial HSP70 ameliorates mitochondrial dysfunction that culminates in irreversible visual loss and atrophy of the optic nerve in EAE suggesting that it may be useful to prevent irreversible disability in patients with optic neuritis and multiple sclerosis (MS).
Collapse
Affiliation(s)
- Venu Talla
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, United States
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, United States
| | - Vince Chiodo
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - John Guy
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
72
|
Wong WK, Cheung AWS, Yu SW, Sha O, Cho EYP. Hepatocyte growth factor promotes long-term survival and axonal regeneration of retinal ganglion cells after optic nerve injury: comparison with CNTF and BDNF. CNS Neurosci Ther 2014; 20:916-29. [PMID: 24992648 DOI: 10.1111/cns.12304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 06/02/2014] [Accepted: 06/04/2014] [Indexed: 11/29/2022] Open
Abstract
AIMS Different trophic factors are known to promote retinal ganglion cell survival and regeneration, but each had their own limitations. We report that hepatocyte growth factor (HGF) confers distinct advantages in supporting ganglion cell survival and axonal regeneration, when compared to two well-established trophic factors ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF). METHODS Ganglion cells in adult hamster were injured by cutting the optic nerve. HGF, CNTF, or BDNF was injected at different dosages intravitreally after injury. Ganglion cell survival was quantified at 7, 14, or 28 days postinjury. Peripheral nerve (PN) grafting to the cut optic nerve of the growth factor-injected eye was performed either immediately after injury or delayed until 7 days post-injury. Expression of heat-shock protein 27 and changes in microglia numbers were quantified in different growth factor groups. The cellular distribution of c-Met in the retina was examined by anti-c-Met immunostaining. RESULTS Hepatocyte Growth Factor (HGF) was equally potent as BDNF in promoting short-term survival (up to 14 days post-injury) and also supported survival at 28 days post-injury when ganglion cells treated by CNTF or BDNF failed to be sustained. When grafting was performed without delay, HGF stimulated twice the number of axons to regenerate compared with control but was less potent than CNTF. However, in PN grafting delayed for 7 days after optic nerve injury, HGF maintained a better propensity of ganglion cells to regenerate than CNTF. Unlike CNTF, HGF application did not increase HSP27 expression in ganglion cells. Microglia proliferation was prolonged in HGF-treated retinas compared with CNTF or BDNF. C-Met was localized to both ganglion cells and Muller cells, suggesting HGF could be neuroprotective via interacting with both neurons and glia. CONCLUSION Compared with CNTF or BDNF, HGF is advantageous in sustaining long-term ganglion cell survival and their propensity to respond to favorable stimuli.
Collapse
Affiliation(s)
- Wai-Kai Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | | | |
Collapse
|
73
|
Kim M, Kim SO, Lee M, Park Y, Kim D, Cho KH, Kim SY, Lee EH. Effects of ginsenoside Rb1 on the stress-induced changes of BDNF and HSP70 expression in rat hippocampus. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:257-262. [PMID: 24975446 DOI: 10.1016/j.etap.2014.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 06/06/2014] [Accepted: 06/11/2014] [Indexed: 06/03/2023]
Abstract
Ginsenoside Rb1 (GRb1) has been determined to exert diverse neuromodulatory effects including antistress effects in the brain. The hippocampus is a key brain structure for memory, learning, and cognition and is especially vulnerable to neurotoxic effects associated with stress. The aim of this study was to further explore neuroprotective potential of GRb1 on stress-mediated changes in hippocampal gene expression. Recent studies recognize agents that inducing brain-derived neurotrophic factor (BDNF) and heat shock protein (HSP) 70 as important neuroprotective approaches. Thus, we specifically determined the effects of GRb1 on mRNA expression of BDNF and HSP70, in a model of immobilization stress. In agreement with these reports, acute immobilization stress led to a decrease and an increase in the mRNA levels of the BDNF and HSP70, respectively, in the hippocampus. When pretreated orally, GRb1 significantly inhibited the stress-mediated decline of BDNF level whereas it further increased the stress-mediated elevation of HSP70 level. Our results strongly suggest GRb1 effective in controlling stress-related hippocampal dysfunction. Our finding also contributes further understanding of medicinal usefulness of GRb1 targeting hippocampal network alteration which is commonly observed in aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Mia Kim
- Department of Cardiovascular & Neurologic Diseases (Stroke Center), Hospital of Oriental Medicine, Kyung Hee University, Seoul 130-702, Republic of Korea
| | - Sung-Ok Kim
- College of Oriental Medicine, Daegu Haany University, Daegu 706-060, Republic of Korea
| | - Moonsung Lee
- Department of East-West Medical Science, Kyung Hee University, Yongin-si 446-701, Republic of Korea
| | - Yeri Park
- Department of East-West Medical Science, Kyung Hee University, Yongin-si 446-701, Republic of Korea
| | - Danhyo Kim
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 446-701, Republic of Korea
| | - Ki-Ho Cho
- Department of Cardiovascular & Neurologic Diseases (Stroke Center), Hospital of Oriental Medicine, Kyung Hee University, Seoul 130-702, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, Incheon 406-799, Republic of Korea
| | - Eunjoo H Lee
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 446-701, Republic of Korea.
| |
Collapse
|
74
|
Choi JI, Kim SD, Kim SH, Lim DJ, Ha SK. Semi-quantitative analyses of hippocampal heat shock protein-70 expression based on the duration of ischemia and the volume of cerebral infarction in mice. J Korean Neurosurg Soc 2014; 55:307-12. [PMID: 25237425 PMCID: PMC4166325 DOI: 10.3340/jkns.2014.55.6.307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/19/2014] [Accepted: 06/11/2014] [Indexed: 11/27/2022] Open
Abstract
Objective We investigated the expression of hippocampal heat shock protein 70 (HSP-70) infarction volume after different durations of experimental ischemic stroke in mice. Methods Focal cerebral ischemia was induced in mice by occluding the middle cerebral artery with the modified intraluminal filament technique. Twenty-four hours after ischemia induction, both hippocampi were extracted for HSP-70 protein analyses. Slices from each hemisphere were stained with 2,3,5-triphenyltetrazolium chloride (2%), and infarction volumes were calculated. HSP-70 levels were evaluated using western blot and enzyme-linked immunosorbent assay (ELISA). HSP-70 subtype (hsp70.1, hspa1a, hspa1b) mRNA levels in the hippocampus were measured using reverse transcription-polymerase chain reaction (RT-PCR). Results Cerebral infarctions were found ipsilateral to the occlusion in 10 mice exposed to transient ischemia (5 each in the 30-min and 60-min occlusion groups), whereas no focal infarctions were noted in any of the sham mice. The average infarct volumes of the 2 ischemic groups were 22.28±7.31 mm3 [30-min group±standard deviation (SD)] and 38.06±9.53 mm3 (60-min group±SD). Western blot analyses and ELISA showed that HSP-70 in hippocampal tissues increased in the infarction groups than in the sham group. However, differences in HSP-70 levels between the 2 infarction groups were statistically insignificant. Moreover, RT-PCR results demonstrated no relationship between the mRNA expression of HSP-70 subtypes and occlusion time or infarction volume. Conclusion Our results indicated no significant difference in HSP-70 expression between the 30- and 60-min occlusion groups despite the statistical difference in infarction volumes. Furthermore, HSP-70 subtype mRNA expression was independent of both occlusion duration and cerebral infarction volume.
Collapse
Affiliation(s)
- Jong-Il Choi
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Sang-Dae Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Se-Hoon Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Dong-Jun Lim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| | - Sung-Kon Ha
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Ansan, Korea
| |
Collapse
|
75
|
Khalouei S, Chow AM, Brown IR. Stress-induced localization of HSPA6 (HSP70B') and HSPA1A (HSP70-1) proteins to centrioles in human neuronal cells. Cell Stress Chaperones 2014; 19:321-7. [PMID: 24061851 PMCID: PMC3982026 DOI: 10.1007/s12192-013-0459-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/14/2013] [Accepted: 08/15/2013] [Indexed: 12/21/2022] Open
Abstract
The localization of yellow fluorescent protein (YFP)-tagged HSP70 proteins was employed to identify stress-sensitive sites in human neurons following temperature elevation. Stable lines of human SH-SY5Y neuronal cells were established that expressed YFP-tagged protein products of the human inducible HSP70 genes HSPA6 (HSP70B') and HSPA1A (HSP70-1). Following a brief period of thermal stress, YFP-tagged HSPA6 and HSPA1A rapidly appeared at centrioles in the cytoplasm of human neuronal cells, with HSPA6 demonstrating a more prolonged signal compared to HSPA1A. Each centriole is composed of a distal end and a proximal end, the latter linking the centriole doublet. The YFP-tagged HSP70 proteins targeted the proximal end of centrioles (identified by γ-tubulin marker) rather than the distal end (centrin marker). Centrioles play key roles in cellular polarity and migration during neuronal differentiation. The proximal end of the centriole, which is involved in centriole stabilization, may be stress-sensitive in post-mitotic, differentiating human neurons.
Collapse
Affiliation(s)
- Sam Khalouei
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, M1C 1A4 ON Canada
| | - Ari M. Chow
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, M1C 1A4 ON Canada
| | - Ian R. Brown
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, M1C 1A4 ON Canada
| |
Collapse
|
76
|
Jing X, Shi Q, Bi W, Zeng Z, Liang Y, Wu X, Xiao S, Liu J, Yang L, Tao E. Rifampicin protects PC12 cells from rotenone-induced cytotoxicity by activating GRP78 via PERK-eIF2α-ATF4 pathway. PLoS One 2014; 9:e92110. [PMID: 24638036 PMCID: PMC3956889 DOI: 10.1371/journal.pone.0092110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/17/2014] [Indexed: 11/30/2022] Open
Abstract
Rifampicin has been proposed as a therapeutic candidate for Parkinson's disease (PD). We previously showed that rifampicin was neuroprotective in PD models in vivo and in vitro. However, the molecular mechanisms underlying are not fully elucidated. In this study, using the comprehensive proteomic analysis, we identified that the 78 kDa glucose-regulated protein (GRP78), a hallmark of the unfolded protein response (UPR), was upregulated in rifampicin-treated PC12 cells. Western blot analysis confirmed GRP78 activation. GRP78 functions cytoprotectively in stressed cells, therefore, we hypothesized that GRP78 mediated rifampicin-induced neuroprotection. Using RNA interference, we found that GRP78 gene knockdown significantly attenuated the neuroprotective effects of rifampicin. Next, we examined three UPR transducers, namely, protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol requiring kinase α (IREα) and activating transcription factor 6 (ATF 6), and how they regulated rifampicin-stimulated GRP78 expression. Our results showed that PERK, eukaryotic initiation factor 2α (eIF2α), and activating transcription factor 4 (ATF4) were activated in rifampicin-treated PC12 cells. Silencing the ATF4 gene using RNAi inhibited GRP78 stimulation. Interestingly, we did not detect significant IREα activation, X-box binding protein 1 mRNA splicing, or ATF6 cleavage up to 24 h after rifampicin treatment. Taken together, our data suggested that rifampicin induced GRP78 via the PERK-eIF2α-ATF4 pathway to protect neurons against rotenone-induced cell damage. Targeting molecules in this pathway could be a novel therapeutic approach for PD treatment.
Collapse
Affiliation(s)
- Xiuna Jing
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qiaoyun Shi
- Department of Radiology, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Wei Bi
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Zhifen Zeng
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanran Liang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xia Wu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Enxiang Tao
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
77
|
Alani B, Salehi R, Sadeghi P, Zare M, Khodagholi F, Arefian E, Hakemi MG, Digaleh H. Silencing of Hsp90 chaperone expression protects against 6-hydroxydopamine toxicity in PC12 cells. J Mol Neurosci 2014; 52:392-402. [PMID: 24234033 DOI: 10.1007/s12031-013-0163-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 10/23/2013] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder that has been shown to be associated with oxidative stress. This phenomenon occurs primarily via generation of 6-hydroxydopamine(6-OHDA) in catecholaminergic neurons leading to activation of apoptosis. The 90-kDa heat shock protein (Hsp90) functions as a chaperone in maintaining the functional stability and viability of cells under a transforming pressure. Since Hsp90 binds to inactive transcription factor heat shock factor-1 (HSF-1), inhibition of Hsp90 could activate HSF-1 and transcription of heat shock element containing genes subsequently, like Hsp70 as an anti-apoptotic factor. Our trial of silencing Hsp90 expression through transfection of Hsp90 siRNAs into neuronal PC12 cells being exposed to 6-OHDA resulted in the inhibition of pro-apoptotic factors, Bax, caspase-3, and PARP and upregulation of anti-apoptotic factor, Bcl2. In this manner,our data suggest a protective role for Hsp70 as it was observed to be induced upon Hsp90 knockdown. Furthermore, our results showed that Hsp90 silencing against 6-OHDA-induced oxidative stress may associate with upregulation of nuclear factor-erythroid 2-related factor 2. In summary, we found that silencing of Hsp90 expression leads to induction of cytoprotective pathways which can protect neurons against apoptosis in a PD model.
Collapse
|
78
|
Kowalczyk M, Owczarek A, Suchanek R, Paul-Samojedny M, Fila-Danilow A, Borkowska P, Kucia K, Kowalski J. Heat shock protein 70 gene polymorphisms are associated with paranoid schizophrenia in the Polish population. Cell Stress Chaperones 2014; 19:205-15. [PMID: 23893339 PMCID: PMC3933617 DOI: 10.1007/s12192-013-0446-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 07/01/2013] [Accepted: 07/10/2013] [Indexed: 01/01/2023] Open
Abstract
HSP70 genes have been considered as promising schizophrenia candidate genes based on their protective role in the central nervous system under stress conditions. In this study, we analyzed the potential implication of HSPA1A +190G/C, HSPA1B +1267A/G, and HSPA1L +2437T/C polymorphisms in the susceptibility to paranoid schizophrenia in a homogenous Caucasian Polish population. In addition, we investigated the association of the polymorphisms with the clinical variables of the disease. Two hundred and three patients with paranoid schizophrenia and 243 healthy controls were enrolled in the study. Polymorphisms of HSPA1A, -1B, and -1L genes were genotyped using the PCR-RFLP technique. Analyses were conducted in entire groups and in subgroups that were stratified according to gender. There were significant differences in the genotype and allele frequencies of HSPA1A polymorphism between the patients and controls. The +190CC genotype and +190C allele were over-represented in the patients and significantly increased the risk for developing schizophrenia (OR = 3.45 and OR = 1.61, respectively). Interestingly, such a risk was higher for females with the +190CC genotype than for males with the +190CC genotype (OR = 5.78 vs. OR = 2.76). We also identified the CGT haplotype as a risk haplotype for schizophrenia and demonstrated the effects of HSPA1A and HSPA1B genotypes on the psychopathology and age of onset. Our study provided the first evidence that the HSPA1A polymorphism may potentially increase the risk of developing paranoid schizophrenia. Further independent analyses in different populations to evaluate the role of gender are needed to replicate these results.
Collapse
Affiliation(s)
- Malgorzata Kowalczyk
- Department of Medical Genetics, Medical University of Silesia, Ostrogorska 30, 41-200, Sosnowiec, Poland,
| | | | | | | | | | | | | | | |
Collapse
|
79
|
The large conductance, calcium-activated K+ (BK) channel is regulated by cysteine string protein. Sci Rep 2014; 3:2447. [PMID: 23945775 PMCID: PMC3744087 DOI: 10.1038/srep02447] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 07/26/2013] [Indexed: 11/08/2022] Open
Abstract
Large-conductance, calcium-activated-K(+) (BK) channels are widely distributed throughout the nervous system, where they regulate action potential duration and firing frequency, along with presynaptic neurotransmitter release. Our recent efforts to identify chaperones that target neuronal ion channels have revealed cysteine string protein (CSPα) as a key regulator of BK channel expression and current density. CSPα is a vesicle-associated protein and mutations in CSPα cause the hereditary neurodegenerative disorder, adult-onset autosomal dominant neuronal ceroid lipofuscinosis (ANCL). CSPα null mice show 2.5 fold higher BK channel expression compared to wild type mice, which is not seen with other neuronal channels (i.e. Cav2.2, Kv1.1 and Kv1.2). Furthermore, mutations in either CSPα's J domain or cysteine string region markedly increase BK expression and current amplitude. We conclude that CSPα acts to regulate BK channel expression, and consequently CSPα-associated changes in BK activity may contribute to the pathogenesis of neurodegenerative disorders, such as ANCL.
Collapse
|
80
|
Sheppard PW, Sun X, Khammash M, Giffard RG. Overexpression of heat shock protein 72 attenuates NF-κB activation using a combination of regulatory mechanisms in microglia. PLoS Comput Biol 2014; 10:e1003471. [PMID: 24516376 PMCID: PMC3916226 DOI: 10.1371/journal.pcbi.1003471] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 12/30/2013] [Indexed: 12/16/2022] Open
Abstract
Overexpression of the inducible heat shock protein 70, Hsp72, has broadly cytoprotective effects and improves outcome following stroke. A full understanding of how Hsp72 protects cells against injury is elusive, though several distinct mechanisms are implicated. One mechanism is its anti-inflammatory effects. We study the effects of Hsp72 overexpression on activation of the transcription factor NF-κB in microglia combining experimentation and mathematical modeling, using TNFα to stimulate a microglial cell line stably overexpressing Hsp72. We find that Hsp72 overexpression reduces the amount of NF-κB DNA binding activity, activity of the upstream kinase IKK, and amount of IκBα inhibitor phosphorylated following TNFα application. Simulations evaluating several proposed mechanisms suggest that inhibition of IKK activation is an essential component of its regulatory activities. Unexpectedly we find that Hsp72 overexpression reduces the initial amount of the RelA/p65 NF-κB subunit in cells, contributing to the attenuated response. Neither mechanism in isolation, however, is sufficient to attenuate the response, providing evidence that Hsp72 relies upon multiple mechanisms to attenuate NF-κB activation. An additional observation from our study is that the induced expression of IκBα is altered significantly in Hsp72 expressing cells. While the mechanism responsible for this observation is not known, it points to yet another means by which Hsp72 may alter the NF-κB response. This study illustrates the multi-faceted nature of Hsp72 regulation of NF-κB activation in microglia and offers further clues to a novel mechanism by which Hsp72 may protect cells against injury. Inducing heat shock or overexpressing certain heat shock proteins (HSPs) is known to protect against brain injury, such as that resulting from stroke. Understanding the mechanisms underlying protection at the cellular and molecular level is a subject of intense research, as such knowledge may prove beneficial in designing future therapies. Regulation of the activation of the key inflammatory transcription factor Nuclear Factor κB (NF-κB) is believed to be one critical mechanism. However how its activation is altered by Hsp72 remains unresolved. Here we examine NF-κB signaling in microglia cells overexpressing Hsp72, combining experimentation and mathematical modeling. We show that Hsp72 affects signaling using at least two essential and distinct mechanisms: attenuation of upstream kinase (IKK) activity and reduction of steady state NF-κB protein levels. We provide numerical evidence suggesting that neither mechanism in isolation is sufficient to account for the observed signaling. Furthermore, our observations suggest an intriguing additional level of regulation of gene expression and protein synthesis of the IκBα inhibitor, which opens interesting new avenues of research. These results provide novel insight into the mechanisms by which Hsp72 may regulate inflammation and protect brain cells from injury.
Collapse
Affiliation(s)
- Patrick W. Sheppard
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Biosystems Science and Engineering, ETH-Zurich, Basel, Switzerland
- * E-mail:
| | - Xiaoyun Sun
- Department of Anesthesia, Stanford University, Stanford, California, United States of America
| | - Mustafa Khammash
- Department of Biosystems Science and Engineering, ETH-Zurich, Basel, Switzerland
| | - Rona G. Giffard
- Department of Anesthesia, Stanford University, Stanford, California, United States of America
| |
Collapse
|
81
|
Brief maternal separation affects brain α1-adrenoceptors and apoptotic signaling in adult mice. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48:161-9. [PMID: 24128685 DOI: 10.1016/j.pnpbp.2013.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/21/2013] [Accepted: 10/03/2013] [Indexed: 01/24/2023]
Abstract
Exposure to adversity during early life is a risk factor for the development of different mood and psychiatric disorders, including depressive-like behaviors. Here, neonatal mice were temporarily but repeatedly (day 1 to day 13) separated from mothers and placed in a testing environment containing a layer of odorless clean bedding (CB). We assessed in adult animals the impact of this early experience on binding sites and mRNA expression of α1-adrenergic receptor subtypes, heat shock proteins (HSPs) and proapoptotic and antiapoptotic members of the Bcl-2 family proteins in different brain regions involved in processing of olfactory information and rewarding stimuli. We found that repeated exposure to CB experience produced anhedonic-like behavior in terms of reduced saccharin intake and α1-adrenoceptor downregulation in piriform and somatosensory cortices, hippocampus, amygdala and discrete thalamic nuclei. We also found a selective decrease of α1B-adrenoceptor binding sites in the cingulate cortex and hippocampus and an increase of hippocampal α1A and α1B receptor, but not of α1D-adrenoceptor, mRNA levels. Moreover, while a significant decrease of antiapoptotic heat shock proteins Hsp72 and Hsp90 was identified in the prefrontal cortex, a parallel increase of antiapoptotic members of Bcl-2 family proteins was found at the hippocampal level. Together, these data provide evidence that the early exposure to CB experience produced enduring downregulation of α1-adrenoceptors in the prefrontal-limbic forebrain/limbic midbrain network, which plays a key role in the processing of olfactory information and reaction to rewarding stimuli. Finally, these data show that CB experience can "prime" the hippocampal circuitry and promote the expression of antiapoptotic factors that can confer potential neuroprotection to subsequent adversity.
Collapse
|
82
|
Hardas SS, Sultana R, Warrier G, Dan M, Wu P, Grulke EA, Tseng MT, Unrine JM, Graham UM, Yokel RA, Butterfield DA. Rat hippocampal responses up to 90 days after a single nanoceria dose extends a hierarchical oxidative stress model for nanoparticle toxicity. Nanotoxicology 2013; 8 Suppl 1:155-66. [DOI: 10.3109/17435390.2013.868059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | | | - Mo Dan
- Department of Pharmaceutical Sciences and
| | - Peng Wu
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Eric A. Grulke
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Michael T. Tseng
- Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY, USA,
| | - Jason M. Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA,
| | - Uschi M. Graham
- Center for Applied Energy Research, University of Kentucky, Lexington, KY, USA,
| | - Robert A. Yokel
- Department of Pharmaceutical Sciences and
- Graduate Center for Toxicology, University of Kentucky Academic Medical Center, Lexington, KY, USA, and
| | - D. Allan Butterfield
- Department of Chemistry,
- Center of Membrane Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
83
|
Jiang Y, Liu X, Li S, Zhang Y, Piao F, Sun X. Identification of differentially expressed proteins related to organophosphorus-induced delayed neuropathy in the brains of hens. J Appl Toxicol 2013; 34:1352-60. [PMID: 24338829 DOI: 10.1002/jat.2965] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/17/2013] [Accepted: 10/22/2013] [Indexed: 12/17/2022]
Abstract
Some organophosphorus compounds can cause organophosphate-induced delayed neuropathy (OPIDN). Incidents have been documented for decades, however, little is known about which proteins contribute to the initiation, progression and development of OPIDN. In this study, 51 hens were divided into three groups. The tri-ortho-cresyl-phosphate (TOCP) group was treated with 1000 mg kg(-1) TOCP whereas the control group was treated with an equivalent volume of vehicle. The PMSF + TOCP group was treated subcutaneously with 40 mg kg(-1) phenylmethylsulfonyl fluoride (PMSF), followed by 1000 mg kg(-1) TOCP 24 h later. Proteins in the brains of hens were separated by two-dimensional polyacrylamide gel electrophoresis on day 5 after TOCP administration. Mass spectrometry identified eight differentially expressed proteins. Among these proteins, downregulated expression of glutamine synthetase (GS) in the brains of hens after TOCP treatment was further confirmed by real time RT-PCR and ELISA. Moreover, the brains of hens exposed to TOCP exhibited increased levels of glutamate (Glu) and cytosolic calcium concentration ([Ca(2+)](i)), and a decreased level of glutamine (Gln). However, there were no significant differences in GS expression or levels of Glu, Gln, and [Ca(2+)](i) in the brains of hens among the groups on day 21 after TOCP administration. These results indicate that TOCP exposure downregulates GS expression in the brains of hens, and that downregulation of GS is accompanied by increased levels of Glu and [Ca(2+)](i) in the early stage after TOCP administration. It is also suggested that the downregulated expression of GS might be associated with OPIDN through the disruption of homeostasis of the Glu-Gln cycle and [Ca(2+) ](i).
Collapse
Affiliation(s)
- Ying Jiang
- Department of Occupational and Environmental of Health, Dalian Medical University, No. 9 Western Section of Lushun South Road, Dalian, Liaoning, 116044, China
| | | | | | | | | | | |
Collapse
|
84
|
Liu W, Liu K, Tao H, Chen C, Zhang JH, Sun X. Hyperoxia preconditioning: the next frontier in neurology? Neurol Res 2013; 34:415-21. [DOI: 10.1179/1743132812y.0000000034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Wenwu Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Kan Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Hengyi Tao
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Chunhua Chen
- Department of Anatomy and EmbryologyPeking University Health Science Center, Beijing, China
| | - John H Zhang
- Department of AnesthesiologyLoma Linda Medical Center, Loma Linda, CA, USA
| | - Xuejun Sun
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| |
Collapse
|
85
|
TAT-Hsp70 induces neuroprotection against stroke via anti-inflammatory actions providing appropriate cellular microenvironment for transplantation of neural precursor cells. J Cereb Blood Flow Metab 2013; 33:1778-88. [PMID: 23881248 PMCID: PMC3824176 DOI: 10.1038/jcbfm.2013.126] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/25/2013] [Accepted: 06/30/2013] [Indexed: 11/08/2022]
Abstract
Heat-shock protein 70 (Hsp70) protects against cerebral ischemia, which is attributed to its chaperone activity. However, recent reports also describe pro-inflammatory actions of Hsp70 via activation of Toll-like receptors (TLR). Using membrane-permeable transactivator of transcription (TAT)-Hsp70, we analyzed TAT-Hsp70-induced neuroprotection and its underlying mechanism after cerebral ischemia in mice. Infusion of TAT-Hsp70 reduced infarct volume and enhanced blood-brain barrier integrity on day 3 poststroke, when given no later than 12 hours. The latter was associated with reduction of microglial activation, although upregulation of pro-inflammatory TLR-2/4 was observed both in verum and in control animals. Nevertheless, protein abundance and nuclear translocation of downstream nuclear factor kappa B (NF-κB) as well as proteasomal degradation of the NF-κB regulator Ikappa B alpha (IκB-α) were significantly reduced by TAT-Hsp70. TAT-Hsp70-induced neuroprotection and functional recovery were restricted to 4 weeks only. However, TAT-Hsp70 provided an appropriate extracellular milieu for delayed intravenous transplantation of adult neural precursor cells (NPCs). Thus, NPCs that were grafted 28 days poststroke induced long-term neuroprotection for at least 3 months, which was not due to integration of grafted cells but rather due to paracrine effects of transplanted NPCs. Conclusively, TAT-Hsp70 ameliorates postischemic inflammation via proteasome inhibition, thus providing an appropriate extracellular milieu for delayed NPC transplantation and culminating in long-term neuroprotection.
Collapse
|
86
|
Giffard RG, Macario AJL, de Macario EC. The future of molecular chaperones and beyond. J Clin Invest 2013; 123:3206-8. [PMID: 24063055 DOI: 10.1172/jci70799] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protection of hair cells by HSP70 released by supporting cells is reported by May et al. in this issue of the JCI. Their findings suggest a new way to reduce ototoxicity from therapeutic medications and raise larger questions about the role and integration of heat shock proteins in non–cell-autonomous responses to stress. Increasing evidence suggests an important role for extracellular heat shock proteins in both the nervous system and the immune system. The work also suggests that defective chaperones could cause ear disease and supports the potential use of chaperone therapeutics.
Collapse
Affiliation(s)
- Rona G Giffard
- Stanford University School of Medicine, 300 Pasteur Drive, Grant Building S272A, Stanford, California 94305-5117, USA.
| | | | | |
Collapse
|
87
|
Paul S, Mahanta S. Association of heat-shock proteins in various neurodegenerative disorders: is it a master key to open the therapeutic door? Mol Cell Biochem 2013; 386:45-61. [PMID: 24096700 DOI: 10.1007/s11010-013-1844-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 09/26/2013] [Indexed: 12/20/2022]
Abstract
A number of acute and chronic neurodegenerative disorders are caused due to misfolding and aggregation of many intra- and extracellular proteins. Protein misfolding and aggregation processes in cells are strongly regulated by cellular molecular chaperones known as heat-shock proteins (Hsps) that include Hsp60, Hsp70, Hsp40, and Hsp90. Recent studies have shown the evidences that Hsps are colocalized in protein aggregates in Alzheimer's disease (AD), Parkinson's disease (PD), Polyglutamine disease (PGD), Prion disease, and other neurodegenerative disorders. This fact indicates that Hsps might have attempted to prevent aggregate formation in cells and thus to suppress disease conditions. Experimental findings have already established in many cases that selective overexpression of Hsps like Hsp70 and Hsp40 prevented the disease progression in various animal models and cellular models. However, recently, various Hsp modulators like geldanamycin, 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin, and celastrol have shown to up-regulate the expression level of Hsp70 and Hsp40, which in turn triggers the solubilization of diseased protein aggregates. Hsps are, therefore, if appropriately selected, an attractive choice for therapeutic targeting in various kinds of neurodegeneration and hence are expected to have strong potential as therapeutic agents in suppressing or curing AD, PD, PGD, and other devastative neurodegenerative disorders. In the present review, we report the experimental findings that describe the implication of Hsps in the development of neurodegeneration and explore the possibility of how Hsps can be used directly or as a target by other agents to prevent various neurodegeneration through preventing aggregation process and thus reducing the toxicity of the oligomers based on the previous reports.
Collapse
Affiliation(s)
- Subhankar Paul
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India,
| | | |
Collapse
|
88
|
Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol 2013; 11:e1001604. [PMID: 23874151 PMCID: PMC3706306 DOI: 10.1371/journal.pbio.1001604] [Citation(s) in RCA: 648] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 05/29/2013] [Indexed: 11/23/2022] Open
Abstract
Neuronal activity provokes myelinating oligodendrocytes to release exosomes by stimulation of ionotropic glutamate receptors, and that once released, these vesicles are internalized by neurons conveying neuroprotection. Reciprocal interactions between neurons and oligodendrocytes are not only crucial for myelination, but also for long-term survival of axons. Degeneration of axons occurs in several human myelin diseases, however the molecular mechanisms of axon-glia communication maintaining axon integrity are poorly understood. Here, we describe the signal-mediated transfer of exosomes from oligodendrocytes to neurons. These endosome-derived vesicles are secreted by oligodendrocytes and carry specific protein and RNA cargo. We show that activity-dependent release of the neurotransmitter glutamate triggers oligodendroglial exosome secretion mediated by Ca2+ entry through oligodendroglial NMDA and AMPA receptors. In turn, neurons internalize the released exosomes by endocytosis. Injection of oligodendroglia-derived exosomes into the mouse brain results in functional retrieval of exosome cargo in neurons. Supply of cultured neurons with oligodendroglial exosomes improves neuronal viability under conditions of cell stress. These findings indicate that oligodendroglial exosomes participate in a novel mode of bidirectional neuron-glia communication contributing to neuronal integrity. Brain function largely depends on the communication between electrically excitable neurons and surrounding glial cells. Myelinating oligodendrocytes are a type of brain cell that insulate major neuronal processes (axons) and help to sustainably maintain axonal health, which is poorly understood in molecular terms. Several cell types release microvesicles termed exosomes that include genetic information (primarily RNA) and can act as vehicles transferring specific cargo to target cells. Here, we demonstrate that exosomes secreted by oligodendrocytes in response to neuronal signals enter neurons to make their cargo functionally available to the neuronal metabolism. We revealed in cultured cells that exosome release from oligodendrocytes is triggered by the neurotransmitter glutamate through activation of ionotropic glutamate receptors. We also show that glial exosomes are internalized by neurons via an endocytic pathway. By modifying oligodendroglial exosomes with a reporter enzyme, we could demonstrate that the exosome cargo is recovered by target neurons in culture as well as in vivo after injection of exosomes into the mouse brain. Neurons challenged with stressful growth conditions were protected when treated with oligodendroglial exosomes. The study introduces a new concept of reciprocal cell communication in the nervous system and identifies the signal-mediated transfer of exosomes from oligodendrocytes to neurons contributing to the preservation of axonal health.
Collapse
|
89
|
Liu X, Sha O, Cho EYP. Remote ischemic postconditioning promotes the survival of retinal ganglion cells after optic nerve injury. J Mol Neurosci 2013; 51:639-46. [PMID: 23733254 DOI: 10.1007/s12031-013-0036-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/20/2013] [Indexed: 02/07/2023]
Abstract
Ischemic conditioning, the application of a mild ischemic stimulus to an ischemia-sensitive structure like the heart or brain either before (preconditioning) or after (postconditioning) its exposure to a lethal ischemic insult, is known to switch on endogenous protective mechanisms. However, most studies of its neuroprotective effect in the central nervous system (CNS) have focused on ischemic damage or related conditions like hypoxia, while its potential in treating other neural diseases remains uncertain. In particular, the recent discovery of remote ischemic postconditioning whereby mild ischemia applied to a region remote from the target after the main ischemic insult also confers protection offers an attractive paradigm to study its potential in other types of neural injury. Retinal ganglion cells damaged by optic nerve transection undergo extensive cell death. However, application of a series of mild ischemic/reperfusion cycles to the hind limb (limb remote ischemic postconditioning) at 10 min or 6 h after optic nerve cut was found to promote ganglion cell survival at 7 days post-injury, with the 10 min postconditioning still exerting protection at 14 days post-injury. Concomitant with the increased ganglion cell survival, 51 % more ganglion cells expressed the small heat shock protein HSP27, when remote ischemic postconditioning was performed at 10 min post-injury, as compared to the sham conditioning group. Our results highlight the potential of using remote ischemic postconditioning as a noninvasive neuroprotective strategy in different CNS disorders like spinal cord and traumatic brain injury.
Collapse
Affiliation(s)
- Xia Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | |
Collapse
|
90
|
Alberi L, Hoey SE, Brai E, Scotti AL, Marathe S. Notch signaling in the brain: in good and bad times. Ageing Res Rev 2013; 12:801-14. [PMID: 23570941 DOI: 10.1016/j.arr.2013.03.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 03/16/2013] [Accepted: 03/22/2013] [Indexed: 01/13/2023]
Abstract
Notch signaling is an evolutionarily conserved pathway, which is fundamental for neuronal development and specification. In the last decade, increasing evidence has pointed out an important role of this pathway beyond embryonic development, indicating that Notch also displays a critical function in the mature brain of vertebrates and invertebrates. This pathway appears to be involved in neural progenitor regulation, neuronal connectivity, synaptic plasticity and learning/memory. In addition, Notch appears to be aberrantly regulated in neurodegenerative diseases, including Alzheimer's disease and ischemic injury. The molecular mechanisms by which Notch displays these functions in the mature brain are not fully understood, but are currently the subject of intense research. In this review, we will discuss old and novel Notch targets and molecular mediators that contribute to Notch function in the mature brain and will summarize recent findings that explore the two facets of Notch signaling in brain physiology and pathology.
Collapse
Affiliation(s)
- Lavinia Alberi
- Unit of Anatomy, Department of Medicine, University of Fribourg, Switzerland.
| | | | | | | | | |
Collapse
|
91
|
Hwang CS, Liu GT, Chang MDT, Liao IL, Chang HT. Elevated serum autoantibody against high mobility group box 1 as a potent surrogate biomarker for amyotrophic lateral sclerosis. Neurobiol Dis 2013; 58:13-8. [PMID: 23639787 DOI: 10.1016/j.nbd.2013.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/20/2013] [Accepted: 04/09/2013] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complicate and progressive onset devastating neurodegenerative disease. Its pathogenic mechanisms remain unclear and there is no specific test for diagnosis. For years, researchers have been vigorously searching for biomarkers associated with ALS to assist clinical diagnosis and monitor disease progression. Some specific inflammatory processes in the central nervous system have been reported to participate in the pathogenesis of ALS. As high mobility group box 1 (HMGB1) is elevated in spinal cord tissues of patients with ALS, we hypothesized, therefore, that serum autoantibody against HMGB1 (HMGB1 autoAb) might represent an effective biomarker for ALS. Patients with ALS, Alzheimer's disease, Parkinson's disease, and healthy age-matched control subjects were recruited for this study. ALS group consisted of 61 subjects, the other groups each consisted of forty subjects. We generated a polyclonal antibody against HMGB1 and developed an ELISA-based methodology for screening serum samples of these subjects. All samples were coded for masked comparison. For statistic analyses, two-tailed Student's t-test, ANOVA, Bonferroni multiple comparison test, Spearman correlation, and receiver operating characteristic curve were applied. We discovered that the level of HMGB1 autoAb significantly increased in patients with ALS as compared with that of patients with Alzheimer's disease, Parkinson's disease, and healthy control subjects. The differences between all groups were robust even at the early stages of ALS progression. More importantly, higher HMGB1 autoAb level was found in more severe disease status with significant correlation. Our study demonstrates that serum HMGB1 autoAb may serve as a biomarker for the diagnosis of ALS and can be used to monitor disease progression.
Collapse
Affiliation(s)
- Chi-Shin Hwang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
| | | | | | | | | |
Collapse
|
92
|
Chow AM, Tang DWF, Hanif A, Brown IR. Induction of heat shock proteins in cerebral cortical cultures by celastrol. Cell Stress Chaperones 2013; 18:155-60. [PMID: 22865541 PMCID: PMC3581628 DOI: 10.1007/s12192-012-0364-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 07/23/2012] [Accepted: 07/24/2012] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis (ALS) are 'protein misfolding disorders' of the mature nervous system that are characterized by the accumulation of protein aggregates and selective cell loss. Different brain regions are impacted, with Alzheimer's affecting cells in the cerebral cortex, Parkinson's targeting dopaminergic cells in the substantia nigra and ALS causing degeneration of cells in the spinal cord. These diseases differ widely in frequency in the human population. Alzheimer's is more frequent than Parkinson's and ALS. Heat shock proteins (Hsps) are 'protein repair agents' that provide a line of defense against misfolded, aggregation-prone proteins. We have suggested that differing levels of constitutively expressed Hsps (Hsc70 and Hsp27) in neural cell populations confer a variable buffering capacity against 'protein misfolding disorders' that correlates with the relative frequencies of these neurodegenerative diseases. The high relative frequency of Alzheimer's may due to low levels of Hsc70 and Hsp27 in affected cell populations that results in a reduced defense capacity against protein misfolding. Here, we demonstrate that celastrol, but not classical heat shock treatment, is effective in inducing a set of neuroprotective Hsps in cultures derived from cerebral cortices, including Hsp70, Hsp27 and Hsp32. This set of Hsps is induced by celastrol at 'days in vitro' (DIV) 13 when cultured cortical cells reached maturity. The inducibility of a set of neuroprotective Hsps in mature cortical cultures at DIV13 suggests that celastrol is a potential agent to counter Alzheimer's disease, a neurodegenerative 'protein misfolding disorder' of the adult brain that targets cells in the cerebral cortex.
Collapse
Affiliation(s)
- Ari M. Chow
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Derek W. F. Tang
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Asad Hanif
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| | - Ian R. Brown
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
| |
Collapse
|
93
|
Eosinophil-derived neurotoxin is elevated in patients with amyotrophic lateral sclerosis. Mediators Inflamm 2013; 2013:421389. [PMID: 23533305 PMCID: PMC3590756 DOI: 10.1155/2013/421389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/24/2012] [Accepted: 01/10/2013] [Indexed: 12/11/2022] Open
Abstract
Background and Objectives. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by loss of motor neurons in the brainstem, motor cortex, and spinal cord. Oxidative stress and neuroinflammation have been implicated in the pathophysiology of ALS. Members of the family of damage-associated molecular patterns, including reactive oxygen species, high-mobility group box 1, and eosinophil-derived neurotoxin (EDN), may participate in pathological conditions. In this study, we aim to discover new biomarker for detecting ALS. Materials and Methods. We examined 44 patients with ALS, 41 patients with Alzheimer's disease, 41 patients with Parkinson's disease, and 44 healthy controls. The concentration of serum EDN was measured using an enzyme-linked immunosorbent assay. Results. EDN levels were significantly increased 2.17-fold in the serum of patients with ALS as compared with healthy controls (P < 0.05). No correlation between the levels of serum EDN and various clinical parameters of ALS was found. Moreover, the levels of serum EDN in patients with Parkinson's disease and Alzheimer's disease and healthy controls were similar. Conclusion. A higher level of serum EDN was found specifically in patients with ALS, indicating that EDN may participate in the pathophysiology of ALS.
Collapse
|
94
|
Coco M, Caggia S, Musumeci G, Perciavalle V, Graziano ACE, Pannuzzo G, Cardile V. Sodium L-lactate differently affects brain-derived neurothrophic factor, inducible nitric oxide synthase, and heat shock protein 70 kDa production in human astrocytes and SH-SY5Y cultures. J Neurosci Res 2013; 91:313-320. [PMID: 23172800 DOI: 10.1002/jnr.23154] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/11/2012] [Accepted: 09/18/2012] [Indexed: 12/12/2022]
Abstract
The present study analyzed the in vitro effects induced by sodium L-lactate on human astrocytes and the SH-SY5Y cell line, when added at concentrations of 5, 10, and 25 mmol/liter. Expression of brain-derived neurotrophic factor (BDNF), inducible nitric oxide synthase (iNOS), and heat shock protein 70 kDa (HSP70) was evaluated by Western blot analysis. Cell viability with MTT, release of nitric oxide (NO) through the Griess reaction, and production of BDNF by enzyme-linked immunoassay was determined. Data indicate that, in SH-SY5Y as well as in cortical astrocytes, after 4 hr sodium L-lactate increases the expression and release of BDNF, iNOS, and NO; after 24 hr, it turns is ineffective for the production of the neurotrophin in SH-SY5Y and not in astrocytes, but the expression of iNOS and release of NO appear to be further increased compared with those after 4 hr. Sodium L-lactate influences differently the expression of HSP70 in SH-SY5Y compared with astrocytes. We propose, based on these findings, that sodium L-lactate affects the expression of BDNF in SH-SY5Y and astrocytes in a different manner: high levels of iNOS and NO expressed in SH-SY5Y have a profound inhibitory effect on the release of BDNF related to a more limited production of HSP70 by SH-SY5Y. In conclusion, the results demonstrate differences in the responses of SH-SY5Y and astrocytes to stimulation by high levels of sodium L-lactate. Sodium L-lactate differently and dose and time dependently influences the expression and release of BDNF, iNOS, NO, and HSP70 depending on the cell type.
Collapse
Affiliation(s)
- Marinella Coco
- Department of Bio-Medical Sciences, Section of Physiology, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
95
|
The Role of HSPA12B in Regulating Neuronal Apoptosis. Neurochem Res 2013; 38:311-20. [DOI: 10.1007/s11064-012-0922-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 10/26/2012] [Accepted: 11/08/2012] [Indexed: 10/27/2022]
|
96
|
Abstract
αB-crystallin is a member of the heat shock protein family that exerts cell protection under several stress-related conditions. Recent studies have revealed that αB-crystallin plays a beneficial role in a mouse model of multiple sclerosis, brain ischemia, and Alexander disease. Whether αB-crystallin plays a role in modulating the secondary damage after CNS trauma is not known. We report here that αB-crystallin mediates protective effects after spinal cord injury. The levels of αB-crystallin are reduced in spinal cord tissue following contusion lesion. In addition, administration of recombinant human αB-crystallin for the first week after contusion injury leads to sustained improvement in locomotor skills and amelioration of secondary tissue damage. We also provide evidence that recombinant human αB-crystallin modulates the inflammatory response in the injured spinal cord, leading to increased infiltration of granulocytes and reduced recruitment of inflammatory macrophages. Furthermore, the delivery of recombinant human αB-crystallin promotes greater locomotor recovery even when the treatment is initiated 6 h after spinal cord injury. Our findings suggest that administration of recombinant human αB-crystallin may be a good therapeutic approach for treating acute spinal cord injury, for which there is currently no effective treatment.
Collapse
|
97
|
Doeppner TR, Ewert TAS, Tönges L, Herz J, Zechariah A, ElAli A, Ludwig AK, Giebel B, Nagel F, Dietz GPH, Weise J, Hermann DM, Bähr M. Transduction of neural precursor cells with TAT-heat shock protein 70 chaperone: therapeutic potential against ischemic stroke after intrastriatal and systemic transplantation. Stem Cells 2012; 30:1297-310. [PMID: 22593021 DOI: 10.1002/stem.1098] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Novel therapeutic concepts against cerebral ischemia focus on cell-based therapies in order to overcome some of the side effects of thrombolytic therapy. However, cell-based therapies are hampered because of restricted understanding regarding optimal cell transplantation routes and due to low survival rates of grafted cells. We therefore transplanted adult green fluorescence protein positive neural precursor cells (NPCs) either intravenously (systemic) or intrastriatally (intracerebrally) 6 hours after stroke in mice. To enhance survival of NPCs, cells were in vitro protein-transduced with TAT-heat shock protein 70 (Hsp70) before transplantation followed by a systematic analysis of brain injury and underlying mechanisms depending on cell delivery routes. Transduction of NPCs with TAT-Hsp70 resulted in increased intracerebral numbers of grafted NPCs after intracerebral but not after systemic transplantation. Whereas systemic delivery of either native or transduced NPCs yielded sustained neuroprotection and induced neurological recovery, only TAT-Hsp70-transduced NPCs prevented secondary neuronal degeneration after intracerebral delivery that was associated with enhanced functional outcome. Furthermore, intracerebral transplantation of TAT-Hsp70-transduced NPCs enhanced postischemic neurogenesis and induced sustained high levels of brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, and vascular endothelial growth factor in vivo. Neuroprotection after intracerebral cell delivery correlated with the amount of surviving NPCs. On the contrary, systemic delivery of NPCs mediated acute neuroprotection via stabilization of the blood-brain-barrier, concomitant with reduced activation of matrix metalloprotease 9 and decreased formation of reactive oxygen species. Our findings imply two different mechanisms of action of intracerebrally and systemically transplanted NPCs, indicating that systemic NPC delivery might be more feasible for translational stroke concepts, lacking a need of in vitro manipulation of NPCs to induce long-term neuroprotection.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany; Department of Neurology, University of Goettingen Medical School, Goettingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Brown IR. CSSI workshop in Brazil highlights "Stress Responses in the Nervous System" in relation to neurodegenerative diseases and neuroprotection. Cell Stress Chaperones 2012; 17:657-60. [PMID: 22886590 PMCID: PMC3468679 DOI: 10.1007/s12192-012-0366-y] [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: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 11/28/2022] Open
Abstract
A review is provided of the two neuroscience sessions entitled "Stress Responses in the Nervous System" that were presented at the ninth Cell Stress Society International Workshop on the "Molecular Biology of the Stress Response" held in Port Alegre, Brazil, May 27-30, 2012. The sessions were organized and chaired by Ian R. Brown (Toronto, Canada) and Maria Estela Andrés (Santiago, Chile).
Collapse
Affiliation(s)
- Ian R Brown
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto-Scarborough, 1265 Military Trail, Toronto, Ontario, Canada.
| |
Collapse
|
99
|
Neuronal hypoxia induces Hsp40-mediated nuclear import of type 3 deiodinase as an adaptive mechanism to reduce cellular metabolism. J Neurosci 2012; 32:8491-500. [PMID: 22723689 DOI: 10.1523/jneurosci.6514-11.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In neurons, the type 3 deiodinase (D3) inactivates thyroid hormone and reduces oxygen consumption, thus creating a state of cell-specific hypothyroidism. Here we show that hypoxia leads to nuclear import of D3 in neurons, without which thyroid hormone signaling and metabolism cannot be reduced. After unilateral hypoxia in the rat brain, D3 protein level is increased predominantly in the nucleus of the neurons in the pyramidal and granular ipsilateral layers, as well as in the hilus of the dentate gyrus of the hippocampal formation. In hippocampal neurons in culture as well as in a human neuroblastoma cell line (SK-N-AS), a 24 h hypoxia period redirects active D3 from the endoplasmic reticulum to the nucleus via the cochaperone Hsp40 pathway. Preventing nuclear D3 import by Hsp40 knockdown resulted an almost doubling in the thyroid hormone-dependent glycolytic rate and quadrupling the transcription of thyroid hormone target gene ENPP2. In contrast, Hsp40 overexpression increased nuclear import of D3 and minimized thyroid hormone effects in cell metabolism. In conclusion, ischemia/hypoxia induces an Hsp40-mediated translocation of D3 to the nucleus, facilitating thyroid hormone inactivation proximal to the thyroid hormone receptors. This adaptation decreases thyroid hormone signaling and may function to reduce ischemia-induced hypoxic brain damage.
Collapse
|
100
|
Meriin AB, Mense M, Colbert JD, Liang F, Bihler H, Zaarur N, Rock KL, Sherman MY. A novel approach to recovery of function of mutant proteins by slowing down translation. J Biol Chem 2012; 287:34264-72. [PMID: 22902621 DOI: 10.1074/jbc.m112.397307] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein homeostasis depends on a balance of translation, folding, and degradation. Here, we demonstrate that mild inhibition of translation results in a dramatic and disproportional reduction in production of misfolded polypeptides in mammalian cells, suggesting an improved folding of newly synthesized proteins. Indeed, inhibition of translation elongation, which slightly attenuated levels of a copepod GFP mutant protein, significantly enhanced its function. In contrast, inhibition of translation initiation had minimal effects on copepod GFP folding. On the other hand, mild suppression of either translation elongation or initiation corrected folding defects of the disease-associated cystic fibrosis transmembrane conductance regulator mutant F508del. We propose that modulation of translation can be used as a novel approach to improve overall proteostasis in mammalian cells, as well as functions of disease-associated mutant proteins with folding deficiencies.
Collapse
Affiliation(s)
- Anatoli B Meriin
- Department of Biochemistry, Boston University Medical School, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|