1
|
Zhao A, Zhang G, Wei H, Yan X, Gan J, Jiang X. Heat shock proteins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic implications. Exp Neurol 2025; 390:115284. [PMID: 40318821 DOI: 10.1016/j.expneurol.2025.115284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) remains a significant challenge in ischemic stroke treatment. Heat shock proteins (HSPs), a cadre of molecular chaperones, have emerged as pivotal regulators in this pathological cascade. This review synthesizes the latest research on HSPs in CIRI from 2013 to 2024 focusing on their multifaceted roles and therapeutic potential. We explore the diverse cellular functions of HSPs, including regulation of oxidative stress, apoptosis, necroptosis, ferroptosis, autophagy, neuroinflammation, and blood-brain barrier integrity. Key HSPs, such as HSP90, HSP70, HSP32, HSP60, HSP47, and small HSPs, are investigated for their specific mechanisms of action in CIRI. Potential therapeutic strategies targeting HSPs, including HSP inhibitors, traditional Chinese medicine components, and gene therapy, are discussed. This review provides a comprehensive understanding of HSPs in CIRI and offers insights into the development of innovative neuroprotective treatments.
Collapse
Affiliation(s)
- Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Hypoxia Tolerant Species: The Wisdom of Nature Translated into Targets for Stroke Therapy. Int J Mol Sci 2021; 22:ijms222011131. [PMID: 34681788 PMCID: PMC8537001 DOI: 10.3390/ijms222011131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Human neurons rapidly die after ischemia and current therapies for stroke management are limited to restoration of blood flow to prevent further brain damage. Thrombolytics and mechanical thrombectomy are the available reperfusion treatments, but most of the patients remain untreated. Neuroprotective therapies focused on treating the pathogenic cascade of the disease have widely failed. However, many animal species demonstrate that neurons can survive the lack of oxygen for extended periods of time. Here, we reviewed the physiological and molecular pathways inherent to tolerant species that have been described to contribute to hypoxia tolerance. Among them, Foxo3 and Eif5A were reported to mediate anoxic survival in Drosophila and Caenorhabditis elegans, respectively, and those results were confirmed in experimental models of stroke. In humans however, the multiple mechanisms involved in brain cell death after a stroke causes translation difficulties to arise making necessary a timely and coordinated control of the pathological changes. We propose here that, if we were able to plagiarize such natural hypoxia tolerance through drugs combined in a pharmacological cocktail it would open new therapeutic opportunities for stroke and likely, for other hypoxic conditions.
Collapse
|
3
|
Vahidinia Z, Mahdavi E, Talaei SA, Naderian H, Tamtaji A, Haddad Kashani H, Beyer C, Azami Tameh A. The effect of female sex hormones on Hsp27 phosphorylation and histological changes in prefrontal cortex after tMCAO. Pathol Res Pract 2021; 221:153415. [PMID: 33857717 DOI: 10.1016/j.prp.2021.153415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Female sex hormones are protective factors against many neurological disorders such as brain ischemia. Heat shock protein like HSP27 is activated after tissue injury. The main purpose of the present study is to determine the effect of a combined estrogen / progesterone cocktail on the morphology of astrocytes, neurons and Hsp27 phosphorylation after cerebral ischemia. METHODS One hour after the MCAO induction, a single dose of estrogen and progesterone was injected. The infarct volume was calculated by TTC staining 24 h after ischemia. Immunohistochemistry was used to show the effects of estrogen and progesterone on astrocyte and neuron morphology, as well as the Western blot technique used for the quantitation of phosphorylated Hsp27. RESULTS The combined dose of estrogen and progesterone significantly decreased astrocytosis after ischemia and increased neuron survival. There was a large increase in Hsp27 phosphorylation in the penumbra ischemic region after stroke, which was significantly reduced by hormone therapy. CONCLUSION Our results indicate that the neuroprotective effect of neurosteroids in the brain may be due to the modulation of heat shock proteins.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Elham Mahdavi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Homayoun Naderian
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Aboutaleb Tamtaji
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Haddad Kashani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Cai M, Zhu Y, Li Z, Josephs-Spaulding J, Zhou Y, Hu Y, Chen H, Liu Y, He W, Zhang J. Profiling the Gene Expression and DNA Methylation in the Mouse Brain after Ischemic Preconditioning. Neuroscience 2019; 406:249-261. [DOI: 10.1016/j.neuroscience.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
|
5
|
Noguchi K, Ali TFS, Miyoshi J, Orito K, Negoto T, Biswas T, Taira N, Koga R, Okamoto Y, Fujita M, Otsuka M, Morioka M. Neuroprotective effects of a novel carnosine-hydrazide derivative on hippocampal CA1 damage after transient cerebral ischemia. Eur J Med Chem 2018; 163:207-214. [PMID: 30522055 DOI: 10.1016/j.ejmech.2018.11.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Ischemia-reperfusion injuries produce reactive oxygen species that promote the peroxide lipid oxidation process resulting in the production of an endogenic lipid peroxide, 4-hydroxy-trans-2-nonenal (4-HNE), a highly cytotoxic aldehyde that induces cell death. We synthesized a novel 4-HNE scavenger - a carnosine-hydrazide derivative, l-carnosine hydrazide (CNN) - and examined its neuroprotective effect in a model of transient ischemia. PC-12 cells were pre-incubated with various doses (0-50 mmol/L) of CNN for 30 min, followed by incubation with 4-HNE (250 μM). An MTT assay was performed 24 h later to examine cell survival. Transient ischemia was induced by bilateral common carotid artery occlusion (BCCO) in the Mongolian gerbil. Animals were assigned to sham-operated (n = 6), placebo-treated (n = 12), CNN pre-treated (20 mg/kg; n = 12), CNN post-treated (100 mg/kg; n = 11), and histidyl hydrazide (a previously known 4-HNE scavenger) post-treated (100 mg/kg; n = 7) groups. Heat shock protein 70 immunoreactivity in the hippocampal CA1 region was evaluated 24 h later, while delayed neuronal death using 4-HNE staining was evaluated 7 days later. Pre-incubation with 30 mmol/L CNN completely inhibited 4-HNE-induced cell toxicity. CNN prevented delayed neuronal death by >60% in the pre-treated group (p < 0.001) and by >40% in the post-treated group (p < 0.01). Histidyl hydrazide post-treatment elicited no protective effect. CNN pre-treatment resulted in high heat shock protein 70 and low 4-HNE immunoreactivity in CA1 pyramidal neurons. Higher 4-HNE immunoreactivity was also found in the placebo-treated animals than in the CNN pre-treated animals. Our novel compound, CNN, elicited highly effective 4-HNE scavenging activity in vitro. Furthermore, CNN administration both pre- and post-BCCO remarkably reduced delayed neuronal death in the hippocampal CA1 region via its induction of heat shock protein 70 and scavenging of 4-HNE.
Collapse
Affiliation(s)
- Kei Noguchi
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Taha F S Ali
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan; Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Junko Miyoshi
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Kimihiko Orito
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Tetsuya Negoto
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Tanima Biswas
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Naomi Taira
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Ryoko Koga
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Yoshinari Okamoto
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Mikako Fujita
- Research Institute for Drug Discovery, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Department of Bioorganic and Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan.
| |
Collapse
|
6
|
Phosphorylated recombinant HSP27 protects the brain and attenuates blood-brain barrier disruption following stroke in mice receiving intravenous tissue-plasminogen activator. PLoS One 2018; 13:e0198039. [PMID: 29795667 PMCID: PMC5993064 DOI: 10.1371/journal.pone.0198039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 05/11/2018] [Indexed: 01/21/2023] Open
Abstract
Loss of integrity of the blood-brain barrier (BBB) in ischemic stroke victims initiates a devastating cascade of events causing brain damage. Maintaining the BBB is important to preserve brain function in ischemic stroke. Unfortunately, recombinant tissue plasminogen activator (tPA), the only effective fibrinolytic treatment at the acute stage of ischemic stroke, also injures the BBB and increases the risk of brain edema and secondary hemorrhagic transformation. Thus, it is important to identify compounds that maintain BBB integrity in the face of ischemic injury in patients with stroke. We previously demonstrated that intravenously injected phosphorylated recombinant heat shock protein 27 (prHSP27) protects the brains of mice with transient middle cerebral artery occlusion (tMCAO), an animal stroke-model. Here, we determined whether prHSP27, in addition to attenuating brain injury, also decreases BBB damage in hyperglycemic tMCAO mice that had received tPA. After induction of hyperglycemia and tMCAO, we examined 4 treatment groups: 1) bovine serum albumin (BSA), 2) prHSP27, 3) tPA, 4) tPA plus prHSP27. We examined the effects of prHSP27 by comparing the BSA and prHSP27 groups and the tPA and tPA plus prHSP27 groups. Twenty-four hours after injection, prHSP27 reduced infarct volume, brain swelling, neurological deficits, the loss of microvessel proteins and endothelial cell walls, and mortality. It also reduced the rates of hemorrhagic transformation, extravasation of endogenous IgG, and MMP-9 activity, signs of BBB damage. Therefore, prHSP27 injection attenuated brain damage and preserved the BBB in tPA-injected, hyperglycemic tMCAO experimental stroke-model mice, in which the BBB is even more severely damaged than in simple tMCAO mice. The attenuation of brain damage and BBB disruption in the presence of tPA suggests the effectiveness of prHSP27 and tPA as a combination therapy. prHSP27 may be a novel therapeutic agent for ischemic stroke patients whose BBBs are injured following tPA injections.
Collapse
|
7
|
Ondruschka B, Rosinsky F, Trauer H, Schneider E, Dreßler J, Franke H. Drug- and/or trauma-induced hyperthermia? Characterization of HSP70 and myoglobin expression. PLoS One 2018; 13:e0194442. [PMID: 29566034 PMCID: PMC5864017 DOI: 10.1371/journal.pone.0194442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/02/2018] [Indexed: 01/04/2023] Open
Abstract
Introduction Heat shock protein 70 (HSP70) expression could be discussed as an adaption that promotes repair and counteracts cell damage. Myoglobin is released upon muscle damage of several pathways. The purpose of the present study was to determine whether the expression of HSP70 in kidney, heart and brain and of myoglobin in the kidney were associated with the cause of death and the survival times after lethal intoxications with three of the drugs most widely used in our local area (Saxony, Germany) as well as after fatal traumatic brain injury (TBI). Methods We retrospectively collected kidney, heart and brain samples of 50 autopsy cases with toxicological proved lethal intoxication (main drugs methamphetamine, morphine, alcohol), 14 TBI cases and 15 fatalities with acute myocardial injury in age- and gender-matched compilations. Results Our main findings suggest that HSP70 is associated with hyperthermal and other stress factors of most cell populations. HSP70 expressions in kidney and heart muscle are useful for a differentiation between fatal intoxications and cases without toxicological influence (p < 0.05). There were significant differences in the cerebral expression patterns between methamphetamine- and morphine-associated deaths compared to alcohol fatalities (p < 0.05). An intensive staining of HSP70 in the pericontusional zone and the hippocampus after TBI (especially neuronal and vascular) was shown even after short survival times and may be useful as an additional marker in questions of vitality or wound age. A relevant myoglobin decoration of renal tubules was only shown for methamphetamine abuse in the study presented. Conclusion In sum, the immunohistochemical characteristics presented can be supportive for determining final death circumstances and minimal trauma survival times but are not isolated usefully for the detection of drug- or trauma-induced hyperthermia.
Collapse
Affiliation(s)
- Benjamin Ondruschka
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Franziska Rosinsky
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Heiner Trauer
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | - Jan Dreßler
- Institute of Legal Medicine, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
Yamamoto Y, Hosoda K, Imahori T, Tanaka J, Matsuo K, Nakai T, Irino Y, Shinohara M, Sato N, Sasayama T, Tanaka K, Nagashima H, Kohta M, Kohmura E. Pentose phosphate pathway activation via HSP27 phosphorylation by ATM kinase: A putative endogenous antioxidant defense mechanism during cerebral ischemia-reperfusion. Brain Res 2018; 1687:82-94. [PMID: 29510140 DOI: 10.1016/j.brainres.2018.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/24/2018] [Accepted: 03/01/2018] [Indexed: 11/19/2022]
Abstract
Molecular mechanism underlying ischemic stroke remains poorly understood. We previously reported glucose 6-phosphate dehydrogenase (G6PD) activity in pentose phosphate pathway (PPP) is activated via heat shock protein 27 (HSP27) phosphorylation at serine 85 (S85) by ataxia telangiectasia mutated (ATM) kinase during cerebral ischemia. This mechanism seems to be endogenous antioxidative system. To determine whether this system also works during reperfusion, we performed comparative metabolic analysis of reperfusion effect on metabolism in rat cortex using middle cerebral artery occlusion (MCAO). Metabolic profiling using gas-chromatography/mass-spectrometry analysis showed changes in metabolic state that depended on reperfusion time. Enrichment analysis showed PPP was significantly upregulated during ischemia-reperfusion. Significant increases in fructose 6-phosphate and ribulose 5-phosphate after reperfusion also suggested enhancement of PPP. In relation to PPP, ischemia-reperfusion induced an increase of up to 69-fold in HSP27 transcripts after 24-h reperfusion. Immunoblotting showed gradual increase in HSP27 protein and marked increase in HSP27 phosphorylation (S85) that were time-dependent (4.5-fold after 24-h reperfusion). G6PD activity was significantly elevated after 1-h MCAO (20%), reduced after 1-h reperfusion, increased gradually thereafter and significantly elevated after 24-h reperfusion. The NADPH/NAD+ ratio displayed similar increasing pattern. Intracerebroventricular injection of ATM kinase inhibitor (KU-55933) significantly reduced HSP27 phosphorylation and G6PD activity, significantly increased protein carbonyl, and resulted in increase in infarct size (100%) 24-h after reperfusion following 90-min MCAO. Consequently, G6PD activation via HSP27 phosphorylation by ATM kinase may be part of endogenous antioxidant defense neuroprotection mechanism that is activated during ischemia-reperfusion. These findings have important implications for treatment of stroke.
Collapse
Affiliation(s)
- Yusuke Yamamoto
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kohkichi Hosoda
- Department of Neurosurgery, Kobe City Nishi-Kobe Medical Center, 5-7-1, Kojidai, Nishi-ku, Kobe 651-2273, Japan.
| | - Taichiro Imahori
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Jun Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuya Matsuo
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomoaki Nakai
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yasuhiro Irino
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoko Sato
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masaaki Kohta
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Eiji Kohmura
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
9
|
Akhundov AG, Mustafaev NJ, Mekhtiev AA. Role of the brain in adaptation of the common and golden carps to adverse factors of chemical origin. J EVOL BIOCHEM PHYS+ 2018. [DOI: 10.1134/s0022093017060035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Allakhverdiyeva TN, Mekhtiev AA. The preconditioning phenomenon and its mechanisms in the common carp as affected by Actara insecticide. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Expression of Hsp27 and Hsp70 and vacuolization in the pituitary glands in cases of fatal hypothermia. Forensic Sci Med Pathol 2017. [DOI: 10.1007/s12024-017-9884-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Lee BS, Jung E, Lee Y, Chung SH. Hypothermia decreased the expression of heat shock proteins in neonatal rat model of hypoxic ischemic encephalopathy. Cell Stress Chaperones 2017; 22:409-415. [PMID: 28285429 PMCID: PMC5425372 DOI: 10.1007/s12192-017-0782-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/26/2017] [Accepted: 02/23/2017] [Indexed: 12/16/2022] Open
Abstract
Hypothermia (HT) is a well-established neuroprotective strategy against neonatal hypoxic ischemic encephalopathy (HIE). The overexpression of heat shock proteins (HSP) has been shown to provide neuroprotection in animal models of stroke. We aimed to investigate the effect of HT on HSP70 and HSP27 expression in a neonatal rat model of HIE. Seven-day-old rat pups were exposed to hypoxia for 90 min to establish the Rice-Vannucci model and were assigned to the following four groups: hypoxic injury (HI)-normothermia (NT, 36 °C), HI-HT (30 °C), sham-NT, and sham-HT. After temperature intervention for 24 h, the mRNA and protein expression of HSP70 and HSP27 were measured. The association between HSP expression and brain injury severity was also evaluated. The brain infarct size was significantly smaller in the HI-HT group than in the HI-NT group. The mRNA and protein expression of both HSPs were significantly greater in the two HI groups, compared to those in the two sham groups. Moreover, among the rat pups subjected to HI, HT significantly reduced the mRNA and protein expression of both HSPs. The mRNA expression level of the HSPs was proportional to the brain injury severity. Post-ischemic HT, i.e., a cold shock attenuated the expression of HSP70 and HSP27 in a neonatal rat model of HIE. Our study suggests that neither HSP70 nor HSP27 expression is involved in the neuroprotective mechanism through which prolonged HT protects against neonatal HIE.
Collapse
Affiliation(s)
- Byong Sop Lee
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 138-736, South Korea.
| | - Euiseok Jung
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 138-736, South Korea
| | - Yeonjoo Lee
- Medical School, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung-Hoon Chung
- Department of Pediatrics, Kyung Hee University School of Medicine, Seoul, South Korea
| |
Collapse
|
13
|
Imahori T, Hosoda K, Nakai T, Yamamoto Y, Irino Y, Shinohara M, Sato N, Sasayama T, Tanaka K, Nagashima H, Kohta M, Kohmura E. Combined metabolic and transcriptional profiling identifies pentose phosphate pathway activation by HSP27 phosphorylation during cerebral ischemia. Neuroscience 2017; 349:1-16. [PMID: 28257891 DOI: 10.1016/j.neuroscience.2017.02.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/10/2017] [Accepted: 02/17/2017] [Indexed: 12/28/2022]
Abstract
The metabolic pathophysiology underlying ischemic stroke remains poorly understood. To gain insight into these mechanisms, we performed a comparative metabolic and transcriptional analysis of the effects of cerebral ischemia on the metabolism of the cerebral cortex using middle cerebral artery occlusion (MCAO) rat model. Metabolic profiling by gas-chromatography/mass-spectrometry analysis showed clear separation between the ischemia and control group. The decreases of fructose 6-phosphate and ribulose 5-phosphate suggested enhancement of the pentose phosphate pathway (PPP) during cerebral ischemia (120-min MCAO) without reperfusion. Transcriptional profiling by microarray hybridization indicated that the Toll-like receptor and mitogen-activated protein kinase (MAPK) signaling pathways were upregulated during cerebral ischemia without reperfusion. In relation to the PPP, upregulation of heat shock protein 27 (HSP27) was observed in the MAPK signaling pathway and was confirmed through real-time polymerase chain reaction. Immunoblotting showed a slight increase in HSP27 protein expression and a marked increase in HSP27 phosphorylation at serine 85 after 60-min and 120-min MCAO without reperfusion. Corresponding upregulation of glucose 6-phosphate dehydrogenase (G6PD) activity and an increase in the NADPH/NAD+ ratio were also observed after 120-min MCAO. Furthermore, intracerebroventricular injection of ataxia telangiectasia mutated (ATM) kinase inhibitor (KU-55933) significantly reduced HSP27 phosphorylation and G6PD upregulation after MCAO, but that of protein kinase D inhibitor (CID755673) did not affect HSP27 phosphorylation. Consequently, G6PD activation via ischemia-induced HSP27 phosphorylation by ATM kinase may be part of an endogenous antioxidant defense neuroprotection mechanism during the earliest stages of ischemia. These findings have important therapeutic implications for the treatment of stroke.
Collapse
Affiliation(s)
- Taichiro Imahori
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kohkichi Hosoda
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | - Tomoaki Nakai
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yusuke Yamamoto
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yasuhiro Irino
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masakazu Shinohara
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Division of Epidemiology, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoko Sato
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masaaki Kohta
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Eiji Kohmura
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
14
|
Zhan L, Liu L, Li K, Wu B, Liu D, Liang D, Wen H, Wang Y, Sun W, Liao W, Xu E. Neuroprotection of hypoxic postconditioning against global cerebral ischemia through influencing posttranslational regulations of heat shock protein 27 in adult rats. Brain Pathol 2017; 27:822-838. [PMID: 27936516 DOI: 10.1111/bpa.12472] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022] Open
Abstract
We previously reported that hypoxic postconditioning (HPC) ameliorated hippocampal neuronal death induced by transient global cerebral ischemia (tGCI) in adult rats. However, the mechanism of HPC-induced neuroprotection is still elusive. Notably, heat shock protein 27 (Hsp27) has recently emerged as a potent neuroprotectant in cerebral ischemia. Although its robust protective effect on stroke has been recognized, the mechanism of Hsp27-mediated neuroprotection is largely unknown. Here, we investigated the potential molecular mechanism by which HPC modulates the posttranslational regulations of Hsp27 after tGCI. We found that HPC increased expression of Hsp27 in CA1 subregion after tGCI. Inhibition of Hsp27 expression with lentivirus-mediated short hairpin RNA (shRNA) abolished the neuroprotection induced by HPC in vivo. Furthermore, pretreatment with cycloheximide, a protein synthesis inhibitor, resulted in a significant decrease in the degradation rate of Hsp27 protein in postconditioned rats, suggesting that the increase in the expression of Hsp27 after HPC might result from its decreased degradation. Next, pretreatment with leupeptin, a lysosomal inhibitor, resulted in an accumulation of Hsp27 after tGCI, indicating that autophagic pathway may be responsible for the degradation of Hsp27. We further showed that the formation of LC3-II and autophagosomes increased after tGCI. Meanwhile, the degradation of Hsp27 was suppressed and neuronal damage was reduced when blocking autophagy with 3-Methyladenine, whereas activating autophagy with rapamycin showed an opposite tendency. Lastly, we confirmed that HPC increased the expression of phosphorylated MAPKAP kinase 2 (MK2) and Hsp27 after tGCI. Also, administration of SB203580, a p38 mitogen-activated protein kinase inhibitor, decreased the expressions of phosphorylated MK2 and Hsp27. Our results suggested that inhibition of Hsp27 degradation mediated by down-regulation of autophagy may induce ischemic tolerance after HPC. Additionally, phosphorylation of Hsp27 induced by MK2 might be associated with the neuroprotection of HPC.
Collapse
Affiliation(s)
- Lixuan Zhan
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Liu Liu
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Kongping Li
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Baoxing Wu
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Dandan Liu
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Donghai Liang
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, 1518 Clifton Road, 2040K, Atlanta, GA, 30322
| | - Haixia Wen
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Yanmei Wang
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Weiwen Sun
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - Weiping Liao
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| | - En Xu
- Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, 510260, China
| |
Collapse
|
15
|
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a disease that occurs when the brain is subjected to hypoxia, resulting in neuronal death and neurological deficits, with a poor prognosis. The mechanisms underlying hypoxic-ischemic brain injury include excitatory amino acid release, cellular proteolysis, reactive oxygen species generation, nitric oxide synthesis, and inflammation. The molecular and cellular changes in HIE include protein misfolding, aggregation, and destruction of organelles. The apoptotic pathways activated by ischemia and hypoxia include the mitochondrial pathway, the extrinsic Fas receptor pathway, and the endoplasmic reticulum stress-induced pathway. Numerous treatments for hypoxic-ischemic brain injury caused by HIE have been developed over the last half century. Hypothermia, xenon gas treatment, the use of melatonin and erythropoietin, and hypoxic-ischemic preconditioning have proven effective in HIE patients. Molecular chaperones are proteins ubiquitously present in both prokaryotes and eukaryotes. A large number of molecular chaperones are induced after brain ischemia and hypoxia, among which the heat shock proteins are the most important. Heat shock proteins not only maintain protein homeostasis; they also exert anti-apoptotic effects. Heat shock proteins maintain protein homeostasis by helping to transport proteins to their target destinations, assisting in the proper folding of newly synthesized polypeptides, regulating the degradation of misfolded proteins, inhibiting the aggregation of proteins, and by controlling the refolding of misfolded proteins. In addition, heat shock proteins exert anti-apoptotic effects by interacting with various signaling pathways to block the activation of downstream effectors in numerous apoptotic pathways, including the intrinsic pathway, the endoplasmic reticulum-stress mediated pathway and the extrinsic Fas receptor pathway. Molecular chaperones play a key role in neuroprotection in HIE. In this review, we provide an overview of the mechanisms of HIE and discuss the various treatment strategies. Given their critical role in the disease, molecular chaperones are promising therapeutic targets for HIE.
Collapse
Affiliation(s)
- Cong Hua
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wei-Na Ju
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hang Jin
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xin Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gang Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
16
|
Nafar F, Williams JB, Mearow KM. Astrocytes release HspB1 in response to amyloid-β exposure in vitro. J Alzheimers Dis 2016; 49:251-63. [PMID: 26444769 DOI: 10.3233/jad-150317] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although heat shock proteins are thought to function primarily as intracellular chaperones, the release and potential extracellular functions of heat shock proteins have been the focus of an increasing number of studies. Our particular interest is HspB1 (Hsp25/27) and as astrocytes are an in vivo source of HspB1 it is a reasonable possibility they could release HspB1 in response to local stresses. Using primary cultures of rat cortical astrocytes, we investigated the extracellular release of HspB1 with exposure to amyloid-β (Aβ). In order to assess potential mechanisms of release, we cotreated the cells with compounds that can modulate protein secretion including Brefeldin A, Methyl β-cyclodextrin, and MAP kinase inhibitors. Exposure to Aβ (0.1, 1.0, 2.0 μM) for 24-48 h resulted in a selective release of HspB1 that was insensitive to BFA treatment; none of the other inhibitors had any detectable influence. Protease protection assays indicated that some of the released HspB1 was associated with a membrane bound fraction, and analysis of exosomal preparations indicated the presence of HspB1 in exosomes. Finally, immunoprecipitation experiments demonstrated that the extracellular HspB1 was able to interact with extracellular Aβ. In summary, Aβ can stimulate release of HspB1 from astrocytes, this release is insensitive to Golgi or lipid raft disruption, and HspB1 can be found either free in the medium or associated with exosomes. This release suggests that there is a potential for extracellular HspB1 to be able to bind and sequester extracellular Aβ.
Collapse
|
17
|
Wang X, Luo Y, Sun H, Feng J, Ma S, Liu J, Huang B. Dynamic expression changes of Bcl-2, Caspase-3 and Hsp70 in middle cerebral artery occlusion rats. Brain Inj 2016; 29:93-7. [PMID: 25158066 DOI: 10.3109/02699052.2014.945958] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND This study aimed to study the dynamic changes of B cell lymphoma/leukaemia 2 (Bcl-2), caspase-3 and heat shock response protein 70 (Hsp70) in blood serum following acute middle cerebral artery occlusion (MCAO) in rats. METHODS Occlusion of the cerebral artery was accomplished via the intraluminal filament, followed by the TTC staining evaluation and neurological deficit score. Meanwhile, the blood serum was extracted at 0.5, 2, 3, 6, 12 and 24 hours and 3 and 7 days after surgery. The serum expression levels of caspase-3, Bcl-2 and Hsp70 were determined using ELISA kits according to the manufacturer's protocols. Expression correlations between Bcl-2 and Hsp70, Bcl-2 and caspase-3 were analysed using correl function. A rats model was successfully established. RESULTS The expression of all three indexes, including Bcl-2, caspase-3 and Hsp70, was significantly increased after surgery (p < 0.05) and peaked at 12, 24 and 24 hours, respectively. Up to 7 days after MCAO, the expression levels of these proteins recovered to the control levels. There were positive correlations between the expressions of Bcl-2 and Hsp70, Bcl-2 and caspase-3 (p < 0.05). CONCLUSIONS The altered expressions of these proteins in the blood serum may result in many symptoms in acute ischaemic stroke individuals.
Collapse
Affiliation(s)
- Xiaoping Wang
- a Department of Neurology , Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital , Chengdu, Sichuan , PR China , and
| | | | | | | | | | | | | |
Collapse
|
18
|
Hao H, Li S, Tang H, Liu B, Cai Y, Shi C, Xiao X. NQDI-1, an inhibitor of ASK1 attenuates acute perinatal hypoxic-ischemic cerebral injury by modulating cell death. Mol Med Rep 2016; 13:4585-92. [PMID: 27081917 PMCID: PMC4878550 DOI: 10.3892/mmr.2016.5123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 03/29/2016] [Indexed: 12/31/2022] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a ubiquitously expressed protein kinase, which regulates cell fate in numerous injury conditions. Therefore, ASK1 may be a promising novel therapeutic target for injury. However, the expression and distribution of ASK1 in the perinatal brain following hypoxia-ischemia (HI) remains to be elucidated. In the present study, western blotting and immunofluorescence were used to determine the expression and distribution of ASK1 and any associated downstream targets in the perinatal rat brain following HI. NQDI‑1, a specific inhibitor of ASK1 was intracerebroventricularly injected following neonatal rats brain insult for neuroprotection. The results revealed an increased expression of ASK1 and this expression was localized to the neurons and astrocytes, compared with the sham controls. Additionally, it was demonstrated that the ASK1/ c‑Jun N‑terminal kinases (JNK) pathway was involved in the brain damage following HI in neonatal rats. Notably, NQDI‑1 significantly inhibited the in vivo expression levels of ASK1, phosphorylated (p‑)JNK, p‑c‑Jun, p53 and caspase 3. Reduced acute hypoxic‑ischemic cerebral injury and cell apoptosis was observed following the injection of NQDI‑1. Collectively, NQDI-1 attenuated acute perinatal hypoxic‑ischemic cerebral injury by inhibiting the expression of ASK1 and cell apoptosis. This may be a promising novel neuroprotective inhibitor for perinatal cerebra injury.
Collapse
Affiliation(s)
- Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Hui Tang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630 P.R. China
| | - Bingqing Liu
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Congcong Shi
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Xin Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
19
|
Dhillon SK, Gunn AJ, Jung Y, Mathai S, Bennet L, Fraser M. Lipopolysaccharide-Induced Preconditioning Attenuates Apoptosis and Differentially Regulates TLR4 and TLR7 Gene Expression after Ischemia in the Preterm Ovine Fetal Brain. Dev Neurosci 2015; 37:497-514. [PMID: 26184807 DOI: 10.1159/000433422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/13/2015] [Indexed: 11/19/2022] Open
Abstract
Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-β mRNA, and increased fetal plasma IFN-β concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-β, suggests the possibility that IFN-β may be an important mediator of endogenous neuroprotection in the developing brain.
Collapse
|
20
|
Kim SH, Lee WS, Lee NM, Chae SA, Yun SW. Neuroprotective effects of mild hypoxia in organotypic hippocampal slice cultures. KOREAN JOURNAL OF PEDIATRICS 2015; 58:142-7. [PMID: 25932036 PMCID: PMC4414629 DOI: 10.3345/kjp.2015.58.4.142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/01/2014] [Accepted: 09/12/2014] [Indexed: 01/18/2023]
Abstract
Purpose The aim of this study was to investigate the potential effects of mild hypoxia in the mature and immature brain. Methods We prepared organotypic slice cultures of the hippocampus and used hippocampal tissue cultures at 7 and 14 days in vitro (DIV) to represent the immature and mature brain, respectively. Tissue cultures were exposed to 10% oxygen for 60 minutes. Twenty-four hours after this hypoxic insult, propidium iodide fluorescence images were obtained, and the damaged areas in the cornu ammonis 1 (CA1), CA3, and dentate gyrus (DG) were measured using image analysis. Results In the 7-DIV group compared to control tissue, hypoxia-exposed tissue showed decreased damage in two regions (CA1: 5.59%±2.99% vs. 4.80%±1.37%, P=0.900; DG: 33.88%±12.53% vs. 15.98%±2.37%, P=0.166), but this decrease was not statistically significant. In the 14-DIV group, hypoxia-exposed tissue showed decreased damage compared to control tissues; this decrease was not significant in the CA3 (24.51%±6.05% vs. 18.31%±3.28%, P=0.373) or DG (15.72%±3.47% vs. 9.91%±2.11%, P=0.134), but was significant in the CA1 (50.91%±5.90% vs. 32.30%±3.34%, P=0.004). Conclusion Although only CA1 tissues cultured for 14 DIV showed significantly less damage after exposure to hypoxia, the other tissues examined in this study showed a tendency towards less damage after hypoxic exposure. Therefore, mild hypoxia might play a protective role in the brain.
Collapse
Affiliation(s)
- Seh Hyun Kim
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
| | - Woo Soon Lee
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
| | - Na Mi Lee
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
| | - Soo Ahn Chae
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sin Weon Yun
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Chidlow G, Wood JPM, Casson RJ. Expression of inducible heat shock proteins Hsp27 and Hsp70 in the visual pathway of rats subjected to various models of retinal ganglion cell injury. PLoS One 2014; 9:e114838. [PMID: 25535743 PMCID: PMC4275305 DOI: 10.1371/journal.pone.0114838] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
Inducible heat shock proteins (Hsps) are upregulated in the central nervous system in response to a wide variety of injuries. Surprisingly, however, no coherent picture has emerged regarding the magnitude, duration and cellular distribution of inducible Hsps in the visual system following injury to retinal ganglion cells (RGCs). The current study sought, therefore, to achieve the following two objectives. The first aim of this study was to systematically characterise the patterns of Hsp27 and −70 expression in the retina and optic nerve in four discrete models of retinal ganglion cell (RGC) degeneration: axonal injury (ON crush), somato-dendritic injury (NMDA-induced excitotoxicity), chronic hypoperfusion (bilateral occlusion of the carotid arteris) and experimental glaucoma. The second aim was to document Hsp27 and −70 expression in the optic tract, the subcortical retinorecipient areas of the brain, and the visual cortex during Wallerian degeneration of RGC axons. Hsp27 was robustly upregulated in the retina in each injury paradigm, with the chronic models, 2VO and experimental glaucoma, displaying a more persistent Hsp27 transcriptional response than the acute models. Hsp27 expression was always associated with astrocytes and with a subset of RGCs in each of the models excluding NMDA. Hsp27 was present within astrocytes of the optic nerve/optic tract in control rats. During Wallerian degeneration, Hsp27 was upregulated in the optic nerve/optic tract and expressed de novo by astrocytes in the lateral geniculate nucleus and the stratum opticum of the superior colliculus. Conversely, the results of our study indicate Hsp70 was minimally induced in any of the models of injury, either in the retina, or in the optic nerve/optic tract, or in the subcortical, retinorecipient areas of the brain. The findings of the present study augment our understanding of the involvement of Hsp27 and Hsp70 in the response of the visual system to RGC degeneration.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
- * E-mail:
| | - John P. M. Wood
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
| | - Robert J. Casson
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
| |
Collapse
|
22
|
Shevtsov MA, Nikolaev BP, Yakovleva LY, Dobrodumov AV, Dayneko AS, Shmonin AA, Vlasov TD, Melnikova EV, Vilisov AD, Guzhova IV, Ischenko AM, Mikhrina AL, Galibin OV, Yakovenko IV, Margulis BA. Neurotherapeutic activity of the recombinant heat shock protein Hsp70 in a model of focal cerebral ischemia in rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:639-50. [PMID: 24920887 PMCID: PMC4044995 DOI: 10.2147/dddt.s62024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recombinant 70 kDa heat shock protein (Hsp70) is an antiapoptotic protein that has a cell protective activity in stress stimuli and thus could be a useful therapeutic agent in the management of patients with acute ischemic stroke. The neuroprotective and neurotherapeutic activity of recombinant Hsp70 was explored in a model of experimental stroke in rats. Ischemia was produced by the occlusion of the middle cerebral artery for 45 minutes. To assess its neuroprotective capacity, Hsp70, at various concentrations, was intravenously injected 20 minutes prior to ischemia. Forty-eight hours after ischemia, rats were sacrificed and brain tissue sections were stained with 2% triphenyl tetrazolium chloride. Preliminary treatment with Hsp70 significantly reduced the ischemic zone (optimal response at 2.5 mg/kg). To assess Hsp70’s neurotherapeutic activity, we intravenously administered Hsp70 via the tail vein 2 hours after reperfusion (2 hours and 45 minutes after ischemia). Rats were then kept alive for 72 hours. The ischemic region was analyzed using a high-field 11 T MRI scanner. Administration of the Hsp70 decreased the infarction zone in a dose-dependent manner with an optimal (threefold) therapeutic response at 5 mg/kg. Long-term treatment of the ischemic rats with Hsp70 formulated in alginate granules with retarded release of protein further reduced the infarct volume in the brain as well as apoptotic area (annexin V staining). Due to its high neurotherapeutic potential, prolonged delivery of Hsp70 could be useful in the management of acute ischemic stroke.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia ; AL Polenov Russian Research Scientific Institute of Neurosurgery, St Petersburg, Russia
| | - Boris P Nikolaev
- Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | | | - Anatolii V Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | - Anastasiy S Dayneko
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Alexey A Shmonin
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia ; Federal Almazov Medical Research Centre, St Petersburg, Russia
| | - Timur D Vlasov
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Elena V Melnikova
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Alexander D Vilisov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), St Petersburg, Russia ; First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Irina V Guzhova
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | | | - Anastasiya L Mikhrina
- IM Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | - Oleg V Galibin
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Igor V Yakovenko
- AL Polenov Russian Research Scientific Institute of Neurosurgery, St Petersburg, Russia
| | - Boris A Margulis
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| |
Collapse
|
23
|
Doberentz E, Genneper L, Böker D, Lignitz E, Madea B. Expression of heat shock proteins (hsp) 27 and 70 in various organ systems in cases of death due to fire. Int J Legal Med 2014; 128:967-78. [PMID: 24740765 DOI: 10.1007/s00414-014-0994-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/31/2014] [Indexed: 10/25/2022]
Abstract
The expression of heat shock proteins (hsp) increases in case of variable types of endogenous and exogenous cellular stress, as for example thermal stress. Immunohistochemical staining with hsp antibodies can visualize these stress proteins. Fifty-three cases of death due to heat and a control group of 100 deaths without any antemortem thermic stress were examined regarding hsp27 and hsp70 expression in myocardial, pulmonary, and renal tissues. The results revealed a correlation between hsp expression, survival time, and cause of death. In cases of death due to fire, the expression of hsp is more extensive than in the control group, especially in pulmonary and renal tissues. The immunohistochemical investigation of an hsp expression can support the proof of vitality in cases of death related to fire.
Collapse
Affiliation(s)
- E Doberentz
- Institute of Legal Medicine, University of Bonn, Stiftsplatz 12, 53111, Bonn, Germany,
| | | | | | | | | |
Collapse
|
24
|
Kwon HM, Kim Y, Yang SI, Kim YJ, Lee SH, Yoon BW. Geldanamycin protects rat brain through overexpression of HSP70 and reducing brain edema after cerebral focal ischemia. Neurol Res 2013; 30:740-5. [DOI: 10.1179/174313208x289615] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
25
|
Human-derived physiological heat shock protein 27 complex protects brain after focal cerebral ischemia in mice. PLoS One 2013; 8:e66001. [PMID: 23785464 PMCID: PMC3681760 DOI: 10.1371/journal.pone.0066001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/30/2013] [Indexed: 02/04/2023] Open
Abstract
Although challenging, neuroprotective therapies for ischemic stroke remain an interesting strategy for countering ischemic injury and suppressing brain tissue damage. Among potential neuroprotective molecules, heat shock protein 27 (HSP27) is a strong cell death suppressor. To assess the neuroprotective effects of HSP27 in a mouse model of transient middle cerebral artery occlusion, we purified a "physiological" HSP27 (hHSP27) from normal human lymphocytes. hHSP27 differed from recombinant HSP27 in that it formed dimeric, tetrameric, and multimeric complexes, was phosphorylated, and contained small amounts of αβ-crystallin and HSP20. Mice received intravenous injections of hHSP27 following focal cerebral ischemia. Infarct volume, neurological deficit scores, physiological parameters, and immunohistochemical analyses were evaluated 24 h after reperfusion. Intravenous injections of hHSP27 1 h after reperfusion significantly reduced infarct size and improved neurological deficits. Injected hHSP27 was localized in neurons on the ischemic side of the brain. hHSP27 suppressed neuronal cell death resulting from cytochrome c-mediated caspase activation, oxidative stress, and inflammatory responses. Recombinant HSP27 (rHSP27), which was artificially expressed and purified from Escherichia coli, and dephosphorylated hHSP27 did not have brain protective effects, suggesting that the phosphorylation of hHSP27 may be important for neuroprotection after ischemic insults. The present study suggests that hHSP27 with posttranslational modifications provided neuroprotection against ischemia/reperfusion injury and that the protection was mediated through the inhibition of apoptosis, oxidative stress, and inflammation. Intravenously injected human HSP27 should be explored for the treatment of acute ischemic strokes.
Collapse
|
26
|
Gao B, Zhang XY, Han R, Zhang TT, Chen C, Qin ZH, Sheng R. The endoplasmic reticulum stress inhibitor salubrinal inhibits the activation of autophagy and neuroprotection induced by brain ischemic preconditioning. Acta Pharmacol Sin 2013; 34:657-66. [PMID: 23603983 DOI: 10.1038/aps.2013.34] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIM To investigate whether endoplasmic reticulum (ER) stress participates in the neuroprotective effects of ischemic preconditioning (IPC)-induced neuroprotection and autophagy activation in rat brains. METHODS The right middle cerebral artery in SD rats was occluded for 10 min to induce focal cerebral IPC, and was occluded permanently 24 h later to induce permanent focal ischemia (PFI). ER stress inhibitor salubrinal (SAL) was injected via intracerebral ventricle infusion 10 min before the onset of IPC. Infarct volume and motor behavior deficits were examined after the ischemic insult. The protein levels of LC3, p62, HSP70, glucose-regulated protein 78 (GRP 78), p-eIF2α and caspase-12 in the ipsilateral cortex were analyzed using immunoblotting. LC3 expression pattern in the sections of ipsilateral cortex was observed with immunofluorescence. RESULTS Pretreatment with SAL (150 pmol) abolished the neuroprotective effects of IPC, as evidenced by the significant increases in mortality, infarct volume and motor deficits after PFI. At the molecular levels, pretreatment with SAL (150 pmol) significantly increased p-eIF2α level, and decreased GRP78 level after PFI, suggesting that SAL effectively inhibited ER stress in the cortex. Furthermore, the pretreatment with SAL blocked the IPC-induced upregulation of LC3-II and downregulation of p62 in the cortex, thus inhibiting the activation of autophagy. Moreover,SAL blocked the upregulation of HSP70, but significantly increased the cleaved caspase-12 level, thus promoting ER stress-dependent apoptotic signaling in the cortex. CONCLUSION ER stress-induced autophagy might contribute to the neuroprotective effect of brain ischemic preconditioning.
Collapse
|
27
|
Abisambra JF, Jinwal UK, Jones JR, Blair LJ, Koren J, Dickey CA. Exploiting the diversity of the heat-shock protein family for primary and secondary tauopathy therapeutics. Curr Neuropharmacol 2012; 9:623-31. [PMID: 22654720 PMCID: PMC3263456 DOI: 10.2174/157015911798376226] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 11/01/2010] [Accepted: 12/06/2010] [Indexed: 12/17/2022] Open
Abstract
The heat shock protein (Hsp) family is an evolutionarily conserved system that is charged with preventing unfolded or misfolded proteins in the cell from aggregating. In Alzheimer’s disease, extracellular accumulation of the amyloid β peptide (Aβ) and intracellular aggregation of the microtubule associated protein tau may result from mechanisms involving chaperone proteins like the Hsps. Due to the ability of Hsps to regulate aberrantly accumulating proteins like Aβ and tau, therapeutic strategies are emerging that target this family of chaperones to modulate their pathobiology. This article focuses on the use of Hsp-based therapeutics for treating primary and secondary tauopathies like Alzheimer’s disease. It will particularly focus on the pharmacological targeting of the Hsp70/90 system and the value of manipulating Hsp27 for treating Alzheimer’s disease.
Collapse
Affiliation(s)
- Jose F Abisambra
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | | | | | | | | | | |
Collapse
|
28
|
Barreto G, White RE, Ouyang Y, Xu L, Giffard RG. Astrocytes: targets for neuroprotection in stroke. Cent Nerv Syst Agents Med Chem 2012; 11:164-73. [PMID: 21521168 DOI: 10.2174/187152411796011303] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/05/2011] [Accepted: 03/09/2011] [Indexed: 02/08/2023]
Abstract
In the past two decades, over 1000 clinical trials have failed to demonstrate a benefit in treating stroke, with the exception of thrombolytics. Although many targets have been pursued, including antioxidants, calcium channel blockers, glutamate receptor blockers, and neurotrophic factors, often the focus has been on neuronal mechanisms of injury. Broader attention to loss and dysfunction of non-neuronal cell types is now required to increase the chance of success. Of the several glial cell types, this review will focus on astrocytes. Astrocytes are the most abundant cell type in the higher mammalian nervous system, and they play key roles in normal CNS physiology and in central nervous system injury and pathology. In the setting of ischemia astrocytes perform multiple functions, some beneficial and some potentially detrimental, making them excellent candidates as therapeutic targets to improve outcome following stroke and in other central nervous system injuries. The older neurocentric view of the central nervous system has changed radically with the growing understanding of the many essential functions of astrocytes. These include K+ buffering, glutamate clearance, brain antioxidant defense, close metabolic coupling with neurons, and modulation of neuronal excitability. In this review, we will focus on those functions of astrocytes that can both protect and endanger neurons, and discuss how manipulating these functions provides a novel and important strategy to enhance neuronal survival and improve outcome following cerebral ischemia.
Collapse
Affiliation(s)
- George Barreto
- Department of Anesthesia, Stanford University School of Medicine, S272, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
29
|
Sheng R, Liu XQ, Zhang LS, Gao B, Han R, Wu YQ, Zhang XY, Qin ZH. Autophagy regulates endoplasmic reticulum stress in ischemic preconditioning. Autophagy 2012; 8:310-25. [PMID: 22361585 DOI: 10.4161/auto.18673] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent studies have suggested that autophagy plays a prosurvival role in ischemic preconditioning (IPC). This study was taken to assess the linkage between autophagy and endoplasmic reticulum (ER) stress during the process of IPC. The effects of IPC on ER stress and neuronal injury were determined by exposure of primary cultured murine cortical neurons to 30 min of OGD 24 h prior to a subsequent lethal OGD. The effects of IPC on ER stress and ischemic brain damage were evaluated in rats by a brief ischemic insult followed by permanent focal ischemia (PFI) 24 h later using the suture occlusion technique. The results showed that both IPC and lethal OGD increased the LC3-II expression and decreased p62 protein levels, but the extent of autophagy activation was varied. IPC treatment ameliorated OGD-induced cell damage in cultured cortical neurons, whereas 3-MA (5-20 mM) and bafilomycin A 1 (75-150 nM) suppressed the neuroprotection induced by IPC. 3-MA, at the dose blocking autophagy, significantly inhibited IPC-induced HSP70, HSP60 and GRP78 upregulation; meanwhile, it also aggregated the ER stress and increased activated caspase-12, caspase-3 and CHOP protein levels both in vitro and in vivo models. The ER stress inhibitor Sal (75 pmol) recovered IPC-induced neuroprotection in the presence of 3-MA. Rapamycin 50-200 nM in vitro and 35 pmol in vivo 24 h before the onset of lethal ischemia reduced ER stress and ischemia-induced neuronal damage. These results demonstrated that pre-activation of autophagy by ischemic preconditioning can boost endogenous defense mechanisms to upregulate molecular chaperones, and hence reduce excessive ER stress during fatal ischemia.
Collapse
Affiliation(s)
- Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Tureyen K, Bowen K, Liang J, Dempsey RJ, Vemuganti R. Exacerbated brain damage, edema and inflammation in type-2 diabetic mice subjected to focal ischemia. J Neurochem 2011; 116:499-507. [PMID: 21133923 DOI: 10.1111/j.1471-4159.2010.07127.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
One of the limiting factors in stroke therapeutic development is the use of animal models that do not well represent the underlying medical conditions of patients. In humans, diabetes increases the risk of stroke incidence as well as post-stroke mortality. To understand the mechanisms that render diabetics to increased brain damage, we evaluated the effect of transient middle cerebral artery occlusion in adult db/db mice. The db/db mouse is a model of type-2 diabetes with four times higher blood sugar than its normoglycemic genetic control(db/+ mouse). Following transient middle cerebral artery occlusion, the db/db mice showed significantly higher mortality, bigger infarcts, increased cerebral edema, worsened neurological status compared to db/+ mice. The db/db mice also showed significantly higher post-ischemic inflammatory markers (ICAM1(+) capillaries, extravasated macrophages/neutrophils and exacerbated proinflammatory gene expression) compared to db/+ mice. In addition, the post-ischemic neuroprotective heat-shock chaperone gene expression was curtailed in the db/db compared to db/+ mice.
Collapse
Affiliation(s)
- Kudret Tureyen
- Department of Neurological Surgery, University of Wisconsin, Madison, WI 53792, USA
| | | | | | | | | |
Collapse
|
31
|
Shintani Y, Terao Y, Ohta H. Molecular mechanisms underlying hypothermia-induced neuroprotection. Stroke Res Treat 2010; 2011:809874. [PMID: 21151700 PMCID: PMC2995905 DOI: 10.4061/2011/809874] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 10/12/2010] [Indexed: 01/25/2023] Open
Abstract
Stroke is a dynamic event in the brain involving heterogeneous cells. There is now compelling clinical evidence that prolonged, moderate cerebral hypothermia initiated within a few hours after severe ischemia can reduce subsequent neuronal death and improve behavioral recovery. The neuroprotective role of hypothermia is also well established in experimental animals. However, the mechanism of hypothermic neuroprotection remains unclear, although, presumably involves the ability of hypothermia to suppress a broad range of injurious factors. In this paper, we addressed this issue by utilizing comprehensive gene and protein expression analyses of ischemic rat brains. To predict precise target molecules, we took advantage of the therapeutic time window and duration of hypothermia necessary to exert neuroprotective effects. We proposed that hypothermia contributes to protect neuroinflammation, and identified candidate molecules such as MIP-3α and Hsp70 that warrant further investigation as targets for therapeutic drugs acting as “hypothermia-like neuroprotectants.”
Collapse
Affiliation(s)
- Yasushi Shintani
- Pharmacology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 2-17-85, Jusohonmachi, Yodogawa-ku, Osaka 532-8686, Japan
| | | | | |
Collapse
|
32
|
Fujiki M, Abe E, Nagai Y, Shiqi K, Kubo T, Ishii K, Abe T, Kobayashi H. Electroconvulsive seizure-induced VEGF is correlated with neuroprotective effects against cerebral infarction: Involvement of the phosphatidylinositol-3 kinase/Akt pathway. Exp Neurol 2010; 225:377-83. [DOI: 10.1016/j.expneurol.2010.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 07/09/2010] [Accepted: 07/13/2010] [Indexed: 11/25/2022]
|
33
|
Zeiger SLH, McKenzie JR, Stankowski JN, Martin JA, Cliffel DE, McLaughlin B. Neuron specific metabolic adaptations following multi-day exposures to oxygen glucose deprivation. Biochim Biophys Acta Mol Basis Dis 2010; 1802:1095-104. [PMID: 20656023 DOI: 10.1016/j.bbadis.2010.07.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/13/2010] [Accepted: 07/19/2010] [Indexed: 12/22/2022]
Abstract
Prior exposure to sub toxic insults can induce a powerful endogenous neuroprotective program known as ischemic preconditioning. Current models typically rely on a single stress episode to induce neuroprotection whereas the clinical reality is that patients may experience multiple transient ischemic attacks (TIAs) prior to suffering a stroke. We sought to develop a neuron-enriched preconditioning model using multiple oxygen glucose deprivation (OGD) episodes to assess the endogenous protective mechanisms neurons implement at the metabolic and cellular level. We found that neurons exposed to a five minute period of glucose deprivation recovered oxygen utilization and lactate production using novel microphysiometry techniques. Using the non-toxic and energetically favorable five minute exposure, we developed a preconditioning paradigm where neurons are exposed to this brief OGD for three consecutive days. These cells experienced a 45% greater survival following an otherwise lethal event and exhibited a longer lasting window of protection in comparison to our previous in vitro preconditioning model using a single stress. As in other models, preconditioned cells exhibited mild caspase activation, an increase in oxidized proteins and a requirement for reactive oxygen species for neuroprotection. Heat shock protein 70 was upregulated during preconditioning, yet the majority of this protein was released extracellularly. We believe coupling this neuron-enriched multi-day model with microphysiometry will allow us to assess neuronal specific real-time metabolic adaptations necessary for preconditioning.
Collapse
|
34
|
Essential role of the redox-sensitive kinase p66shc in determining energetic and oxidative status and cell fate in neuronal preconditioning. J Neurosci 2010; 30:5242-52. [PMID: 20392947 DOI: 10.1523/jneurosci.6366-09.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic preconditioning is a phenomenon in which low-level stressful stimuli upregulate endogenous defensive programs, resulting in subsequent resistance to otherwise lethal injuries. We previously observed that signal transduction systems typically associated with neurodegeneration such as caspase activation are requisite events for the expression of tolerance and induction of HSP70. In this work, we sought to determine the extent and duration of oxidative and energetic dysfunction as well as the role of effector kinases on metabolic function in preconditioned cells. Using an in vitro neuronal culture model, we observed a robust increase in Raf and p66(Shc) activation within 1 h of preconditioning. Total ATP content decreased by 25% 3 h after preconditioning but returned to baseline by 24 h. Use of a free radical spin trap or p66(shc) inhibitor increased ATP content whereas a Raf inhibitor had no effect. Phosphorylated p66(shc) rapidly relocalized to the mitochondria and in the absence of activated p66(shc), autophagic processing increased. The constitutively expressed chaperone HSC70 relocalized to autophagosomes. Preconditioned cells experience significant total oxidative stress measured by F(2)-isoprostanes and neuronal stress evaluated by F(4)-neuroprostane measurement. Neuroprostane levels were enhanced in the presence of Shc inhibitors. Finally, we found that inhibiting either p66(shc) or Raf blocked neuroprotection afforded by preconditioning as well as upregulation of HSP70, suggesting both kinases are critical for preconditioning but function in fundamentally different ways. This is the first work to demonstrate the essential role of p66(shc) in mediating requisite mitochondrial and energetic compensation after preconditioning and suggests a mechanism by which protein and organelle damage mediated by ROS can increase HSP70.
Collapse
|
35
|
The phosphatidylinositol-3 kinase/Akt pathway mediates geranylgeranylacetone-induced neuroprotection against cerebral infarction in rats. Brain Res 2010; 1330:151-7. [PMID: 20206146 DOI: 10.1016/j.brainres.2010.02.074] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 02/21/2010] [Accepted: 02/22/2010] [Indexed: 11/20/2022]
Abstract
Previous studies demonstrated the cytoprotective effect of geranylgeranylacetone (GGA), a heat shock protein inducer, against ischemic insult. Phosphatidylinositol-3 kinase/Akt (PI3K/Akt) is thought to be an important factor that mediates neuroprotection. However, the signaling pathways in the brain in vivo after oral GGA administration remain unclear. We measured and compared infarction volumes to investigate the effect of GGA on cerebral infarction induced by permanent middle cerebral artery occlusion in rats. We evaluated the effects of pretreatment with 5-hydroxydecanoate (5HD), a specific mitochondrial ATP-sensitive potassium (mitoK(ATP)) channel inhibitor; diazoxide (DZX), a selective mitoK(ATP) channel opener and wortmannin (Wort), a specific PI3K inhibitor of GGA-induced neuroprotection against infarction volumes. To clarify the relationship between PI3K/Akt activation and neuroprotection, we used immunoblot analysis to determine the amount of p-Akt proteins present after GGA administration with or without Wort treatment. Neuroprotective effects of GGA (pretreatment with a single oral GGA dose (800 mg/kg) 48 h before ischemia) were prevented by 5HD, DZX and Wort pretreatment, which indicates that the selective mitoK(ATP) channel and the PI3K/Akt pathway may mediate GGA-dependent protection. Oral GGA-induced p-Akt and GGA pretreatment enhanced ischemia-induced p-Akt, both of which were prevented by Wort pretreatment. These results suggest that a single oral dose of GGA induces p-Akt and that GGA plays an important role in neuroprotection against cerebral ischemia through the mitoK(ATP) channel opening.
Collapse
|
36
|
Stetler RA, Gao Y, Signore AP, Cao G, Chen J. HSP27: mechanisms of cellular protection against neuronal injury. Curr Mol Med 2010; 9:863-72. [PMID: 19860665 DOI: 10.2174/156652409789105561] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The heat shock protein (HSP) family has long been associated with a generalized cellular stress response, particularly in terms of recognizing and chaperoning misfolded proteins. While HSPs in general appear to be protective, HSP27 has recently emerged as a particularly potent neuroprotectant in a number of diverse neurological disorders, ranging from ALS to stroke. Although its robust protective effect on a number of insults has been recognized, the mechanisms and regulation of HSP27's protective actions are still undergoing intense investigation. On the basis of recent studies, HSP27 appears to have a dynamic and diverse range of function in cellular survival. This review provides a forum to compare and contrast recent literature exploring the protective mechanism and regulation of HSP27, focusing on neurological disorders in particular, as they represent a range from protein aggregate-associated diseases to acute stress.
Collapse
Affiliation(s)
- R A Stetler
- Department of Neurology, University of Pittsburgh, 507 South Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
37
|
Hou Y, Wei H, Luo Y, Liu G. Modulating expression of brain heat shock proteins by estrogen in ovariectomized mice model of aging. Exp Gerontol 2009; 45:323-30. [PMID: 19836443 DOI: 10.1016/j.exger.2009.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 09/25/2009] [Accepted: 10/09/2009] [Indexed: 11/26/2022]
Abstract
Heat shock proteins (HSPs) serve as molecular chaperones and endogenous cytoprotective factors. Two of the well-studied HSPs, HSP70, and HSP27 can be significantly induced in many areas of brain by a variety of stressors. A decrease in expression of brain HSPs has been documented in aged brain. Estrogen is well known as a neuroprotective hormone, and it has been reported that estrogen can regulate HSP70 and HSP27 expression in neuronal cells. In this study, the relationship between estrogen and heat stress-induced brain HSPs expression in young and aged ovariectomized (OVX) mice was investigated. Our results show that heat stress-induced levels of HSP70 proteins and mRNA transcripts was significantly lower in brain of aged (12 month) OVX mice, compared with young (2 month) OVX mice group. Estrogen supplementation (17beta-estradiol 0.5mg/kg for 7 days) restored heat stress-induced brain HSP70 expression and attenuated heat stress-induced brain DNA fragmentation, caspase 3 activation and mitochondrial leakage of cytochrome c and AIF in OVX mice. These results suggest that estrogen deficiency during aging down-regulates heat stress-induced brain HSP70 expression, which reveals a previously unknown link between estrogen deficiency and stress response elements.
Collapse
Affiliation(s)
- Yan Hou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
38
|
Chitosan prevents oxidative stress-induced amyloid β formation and cytotoxicity in NT2 neurons: involvement of transcription factors Nrf2 and NF-κB. Mol Cell Biochem 2009; 337:39-51. [DOI: 10.1007/s11010-009-0284-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 10/08/2009] [Indexed: 11/26/2022]
|
39
|
Windisch BK, LeVatte TL, Archibald ML, Chauhan BC. Induction of heat shock proteins 27 and 72 in retinal ganglion cells after acute pressure-induced ischaemia. Clin Exp Ophthalmol 2009; 37:299-307. [PMID: 19472539 DOI: 10.1111/j.1442-9071.2009.02032.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND We wanted to investigate whether heat shock protein (HSP) 27 and HSP 72 are induced in retinal ganglion cells (RGCs) after acute intraocular pressure (IOP)-induced ischaemia. METHODS Retinal ischaemia was induced by acutely increasing IOP to 100-110 mmHg for 30 or 90 min unilaterally in Sprague Dawley rats. A fluorescent tracer (fluorogold, FG) was applied to the superior colliculi to label RGCs. Twenty-four hours, 1 week or 2 weeks after of IOP elevation, rats were killed, RGCs counted, and immunohistochemical labelling of the retina was performed. HSP-positive RGCs were counted and normalized HSP RGC counts determined. RESULTS The ratio of FG-positive labelled RGCs in the experimental to the contralateral eye as a marker of RGC survival remained unchanged after 30 min of ischaemia: 1.09 +/- 0.11 at 1 week, and 0.94 +/- 0.28 at 2 weeks. After 90 min of ischaemia RGC survival decreased to 0.19 +/- 0.14 at 1 week, and 0.20 +/- 0.14 at 2 weeks. After 30 min of ischaemia, the normalized HSP 27- and HSP 72-positive RGC count was detected at highest levels (HSP 27: 5.42 +/- 1.18; HSP 72: 12.23 +/- 1.24) at 2 weeks compared with controls,whereas after 90 min ischaemia it was detected at higher levels at 1 week (HSP 27: 52.63 +/- 3.65; HSP 72: 206.84 +/- 60.38), as well as at 2 weeks (HSP 27: 89.00 +/- 17.21; HSP 72: 191.00 +/- 50.05). CONCLUSION These results demonstrate an enhanced induction of HSP 27 and HSP 72 after 90 min acute IOP-induced ischaemia. In contrast to 30 min ischaemia, we showed time-dependent loss of RGCs after 90 min of ischaemia after 1 week or 2 weeks.
Collapse
Affiliation(s)
- Bettina K Windisch
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | |
Collapse
|
40
|
Benardete EA, Bergold PJ. Genomic analysis of ischemic preconditioning in adult rat hippocampal slice cultures. Brain Res 2009; 1292:107-22. [PMID: 19631194 DOI: 10.1016/j.brainres.2009.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 07/04/2009] [Accepted: 07/08/2009] [Indexed: 01/08/2023]
Abstract
Understanding endogenous mechanisms of neuroprotection may have important clinical applications. It is well established that brain tissue becomes more resistant to ischemic injury following a sublethal ischemic insult. This process, called ischemic preconditioning (IPC), can be induced in adult rat hippocampal slice cultures by a brief oxygen-glucose deprivation (OGD) [Hassen, G.W., Tian, D., Ding, D., Bergold, P.J., 2004. A new model of ischemic preconditioning using young adult hippocampal slice cultures. Brain Res. Brain Res. Protoc. 13, 135-143]. We have analyzed the changes in gene expression brought about by IPC in this model in order to understand the mechanisms involved. Total RNA was isolated at different time points following a brief OGD (3, 6 and 12 h) and used to probe genome-wide expression microarrays. Genes were identified that were significantly up- or down-regulated relative to controls. We placed genes that were differentially expressed into statistically significant groups based on Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO) terms. Genes involved in signal transduction, transcription, and oxidative phosphorylation are differentially expressed at each time point. The analysis demonstrates that alterations in signaling pathways (TGF-beta, Wnt, MAPK, ErbB, Toll-like receptor, JAK-STAT, VEGF) consistently accompany IPC. RT-PCR was used to confirm that members of these signaling pathways are regulated as predicted by the microarray analysis. We verified that protein translation following OGD is necessary for IPC. We also found that blocking the NMDA receptor during OGD does not significantly inhibit IPC in this model or produce large changes in gene expression. Our data thus suggests that changes in signaling pathways and their down-stream targets play an important role in triggering endogenous neuroprotection.
Collapse
Affiliation(s)
- Ethan A Benardete
- Department of Neurosurgery, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
41
|
|
42
|
Kalmar B, Greensmith L. Induction of heat shock proteins for protection against oxidative stress. Adv Drug Deliv Rev 2009; 61:310-8. [PMID: 19248813 DOI: 10.1016/j.addr.2009.02.003] [Citation(s) in RCA: 331] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 02/14/2009] [Indexed: 10/21/2022]
Abstract
Heat shock proteins (Hsps) have been studied for many years and there is now a large body of evidence that demonstrates the role of Hsp upregulation in tissue and cell protection in a wide variety of stress conditions. Oxidative stress is known to be involved in a number of pathological conditions, including neurodegeneration, cardiovascular disease and stroke, and even plays a role in natural aging. In this review we summarize the current understanding of the role of Hsps and the heat shock response (HSR) in these pathological conditions and discuss the therapeutic potential of an Hsp therapy for these disorders. However, although an Hsp based therapy appears to be a promising approach for the treatment of diseases that involve oxidative damage, there are some significant hurdles that must be overcome before this approach can be successful. For example, to be effective an Hsp based therapy will need to ensure that the upregulation of Hsps occurs in the right place (i.e. be cell specific), at the right time and to a level and specificity that ensures that all the important binding partners, namely the co-chaperones, are also present at the appropriate levels. It is therefore unlikely that strategies that involve genetic modifications that result in overexpression of specific Hsps will achieve such sophisticated and coordinated effects. Similarly, it is likely that some pharmaceutical inducers of Hsps may be too generic to achieve the desired specific effects on Hsp expression, or may simply fail to reach their target cells due to delivery problems. However, if these difficulties can be overcome, it is clear that an effective Hsp based therapy would be of great benefit to the wide range of depilating conditions in which oxidative stress plays a critical role.
Collapse
|
43
|
Kesaraju S, Schmidt-Kastner R, Prentice HM, Milton SL. Modulation of stress proteins and apoptotic regulators in the anoxia tolerant turtle brain. J Neurochem 2009; 109:1413-26. [PMID: 19476552 DOI: 10.1111/j.1471-4159.2009.06068.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Freshwater turtles survive prolonged anoxia and reoxygenation without overt brain damage by well-described physiological processes, but little work has been done to investigate the molecular changes associated with anoxic survival. We examined stress proteins and apoptotic regulators in the turtle during early (1 h) and long-term anoxia (4, 24 h) and reoxygenation. Western blot analyses showed changes within the first hour of anoxia; multiple stress proteins (Hsp72, Grp94, Hsp60, Hsp27, and HO-1) increased while apoptotic regulators (Bcl-2 and Bax) decreased. Levels of the ER stress protein Grp78 were unchanged. Stress proteins remained elevated in long-term anoxia while the Bcl-2/Bax ratio was unaltered. No changes in cleaved caspase 3 levels were observed during anoxia while apoptosis inducing factor increased significantly. Furthermore, we found no evidence for the anoxic translocation of Bax from the cytosol to mitochondria, nor movement of apoptosis inducing factor between the mitochondria and nucleus. Reoxygenation did not lead to further increases in stress proteins or apoptotic regulators except for HO-1. The apparent protection against cell damage was corroborated with immunohistochemistry, which indicated no overt damage in the turtle brain subjected to anoxia and reoxygenation. The results suggest that molecular adaptations enhance pro-survival mechanisms and suppress apoptotic pathways to confer anoxia tolerance in freshwater turtles.
Collapse
Affiliation(s)
- Shailaja Kesaraju
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431, USA.
| | | | | | | |
Collapse
|
44
|
Abstract
BACKGROUND Investigations following stroke first of all require information about the spatio-temporal dimension of the ischemic core as well as of perilesional and remote affected tissue. Here we systematically evaluated regions differently impaired by focal ischemia. METHODOLOGY/PRINCIPAL FINDINGS Wistar rats underwent a transient 30 or 120 min suture-occlusion of the middle cerebral artery (MCAO) followed by various reperfusion times (2 h, 1 d, 7 d, 30 d) or a permanent MCAO (1 d survival). Brains were characterized by TTC, thionine, and immunohistochemistry using MAP2, HSP72, and HSP27. TTC staining reliably identifies the infarct core at 1 d of reperfusion after 30 min MCAO and at all investigated times following 120 min and permanent MCAO. Nissl histology denotes the infarct core from 2 h up to 30 d after transient as well as permanent MCAO. Absent and attenuated MAP2 staining clearly identifies the infarct core and perilesional affected regions at all investigated times, respectively. HSP72 denotes perilesional areas in a limited post-ischemic time (1 d). HSP27 detects perilesional and remote impaired tissue from post-ischemic day 1 on. Furthermore a simultaneous expression of HSP72 and HSP27 in perilesional neurons was revealed. CONCLUSIONS/SIGNIFICANCE TTC and Nissl staining can be applied to designate the infarct core. MAP2, HSP72, and HSP27 are excellent markers not only to identify perilesional and remote areas but also to discriminate affected neuronal and glial populations. Moreover markers vary in their confinement to different reperfusion times. The extent and consistency of infarcts increase with prolonged occlusion of the MCA. Therefore interindividual infarct dimension should be precisely assessed by the combined use of different markers as described in this study.
Collapse
|
45
|
Meller R. The role of the ubiquitin proteasome system in ischemia and ischemic tolerance. Neuroscientist 2009; 15:243-60. [PMID: 19181875 DOI: 10.1177/1073858408327809] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ubiquitin modification targets a protein for rapid degradation by the proteasome. However, polyubiquitination of proteins can result in multiple functions depending on the topology of the ubiquitin chain. Therefore, ubiquitin signaling offers a more complex and versatile biology compared with many other posttranslational modifications. One area of potential for the application of this knowledge is the field of ischemia-induced brain damage, as occurs following a stroke. The ubiquitin proteasome system may exert a dual role on neuronal outcome following ischemia. Harmful ischemia results in an overload of the ubiquitin proteasome system, and blocking the proteasome reduces brain infarction following ischemia. However, the rapid and selective degradation of proteins following brief ischemia results in endogenous protection against ischemia. Therefore, further understanding of the molecular signaling mechanisms that regulate the ubiquitin proteasome system may reveal novel therapeutic targets to reduce brain damage when ischemia is predicted or reduce the activation of the cell death mechanisms and the inflammatory response following stroke. The aim of this review is to discuss some of the recent advances in the understanding of protein ubiquitination and its implications for novel stroke therapies.
Collapse
Affiliation(s)
- Robert Meller
- Legacy Clinical Research and Technology Center, Portland, Oregon, USA.
| |
Collapse
|
46
|
Stetler RA, Cao G, Gao Y, Zhang F, Wang S, Weng Z, Vosler P, Zhang L, Signore A, Graham SH, Chen J. Hsp27 protects against ischemic brain injury via attenuation of a novel stress-response cascade upstream of mitochondrial cell death signaling. J Neurosci 2008; 28:13038-55. [PMID: 19052195 PMCID: PMC2614130 DOI: 10.1523/jneurosci.4407-08.2008] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 10/15/2008] [Accepted: 10/21/2008] [Indexed: 11/21/2022] Open
Abstract
Heat shock protein 27 (Hsp27), a recently discovered member of the heat shock protein family, is markedly induced in the brain after cerebral ischemia and other injury states. In non-neuronal systems, Hsp27 has potent cell death-suppressing functions. However, the mechanism of Hsp27-mediated neuroprotection has not yet been elucidated. Using transgenic and viral overexpression of Hsp27, we investigated the molecular mechanism by which Hsp27 exerts its neuroprotective effect. Overexpression of Hsp27 conferred long-lasting tissue preservation and neurobehavioral recovery, as measured by infarct volume, sensorimotor function, and cognitive tasks up to 3 weeks following focal cerebral ischemia. Examination of signaling pathways critical to neuronal death demonstrated that Hsp27 overexpression led to the suppression of the MKK4/JNK kinase cascade. While Hsp27 overexpression did not suppress activation of an upstream regulatory kinase of the MKK/JNK cascade, ASK1, Hsp27 effectively inhibited ASK1 activity via a physical association through its N-terminal domain and the kinase domain of ASK1. The N-terminal region of Hsp27 was required for neuroprotective function against in vitro ischemia. Moreover, knockdown of ASK1 or inhibition of the ASK1/MKK4 cascade effectively inhibited cell death following neuronal ischemia. This underscores the importance of this kinase cascade in the progression of ischemic neuronal death. Inhibition of PI3K had no effect on Hsp27-mediated neuroprotection, suggesting that Hsp27 does not promote cell survival via activation of PI3K/Akt. Based on these findings, we conclude that overexpression of Hsp27 confers long-lasting neuroprotection against ischemic brain injury via a previously unexplored association and inhibition of ASK1 kinase signaling.
Collapse
Affiliation(s)
- R. Anne Stetler
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai 200032, China, and
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Guodong Cao
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai 200032, China, and
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai 200032, China, and
| | - Feng Zhang
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Suping Wang
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Zhongfang Weng
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Peter Vosler
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Lili Zhang
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Armando Signore
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Steven H. Graham
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Jun Chen
- Department of Neurology and Center of Cerebrovascular Diseases Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai 200032, China, and
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
47
|
Park MK, Kang YJ, Lee HS, Kim HJ, Seo HG, Lee JH, Chang KC. The obligatory role of COX-2 expression for induction of HO-1 in ischemic preconditioned rat brain. Biochem Biophys Res Commun 2008; 377:1191-4. [DOI: 10.1016/j.bbrc.2008.10.149] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 10/26/2008] [Indexed: 12/20/2022]
|
48
|
Hsu TC, Chen YC, Lai WX, Chiang SY, Huang CY, Tzang BS. Beneficial effects of treatment with cystamine on brain in NZB/W F1 mice. Eur J Pharmacol 2008; 591:307-14. [DOI: 10.1016/j.ejphar.2008.06.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Accepted: 06/23/2008] [Indexed: 11/26/2022]
|
49
|
Sang H, Li J, Liu J, Wang Z, Huo T, Sun J, Xiong L. Preconditioning with +Gz acceleration (head-to-foot inertial load) produces neuroprotection against transient focal cerebral ischemia in rats. Neurosci Lett 2008; 445:78-82. [PMID: 18778750 DOI: 10.1016/j.neulet.2008.08.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 08/18/2008] [Accepted: 08/21/2008] [Indexed: 10/21/2022]
Abstract
Ischemic preconditioning is considered to be the most robust endogenous neuroprotectant. However, the conventional ischemic preconditioning protocol is both invasive and impractical to apply. The aim of the present study was to evaluate whether preconditioning with +Gz centrifuge acceleration (head-to-foot inertial load) which could induce brief episodes of sublethal ischemia in brain had neuroprotection against focal cerebral ischemic injury. A total of 85 male Sprague-Dawley rats were randomly assigned to five groups (n = 17 in each). The 2 Gz, 4 Gz, 6 Gz and 8 Gz groups were subjected to 3 min exposures at +2 Gz, +4 Gz, +6 Gz and +8 Gz, respectively for consecutive three times in animal centrifuge, with a 30-min rest period between each centrifuge run. The control group had no exposure to +Gz acceleration. Twenty-four hours after the last pretreatment, 12 rats in each groups were subjected to focal cerebral ischemia for 120 min and the other five rats in each group were sacrificed to measure the expression of heat shock protein 70(HSP70) in hippocampus by Western blot analysis. The results indicated that the 6 Gz and 8 Gz groups showed smaller infarct volume and lower neurologic deficit scores than the control group. The expression of HSP70 was significantly increased in 6 Gz and 8 Gz groups than those in the control group. Therefore, preconditioning with +Gz acceleration produced delayed neuroprotection against focal cerebral ischemia and that the neuroprotection may be related to the induction of HSP70.
Collapse
Affiliation(s)
- Hanfei Sang
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Zhan X, Kim C, Sharp FR. Very brief focal ischemia simulating transient ischemic attacks (TIAs) can injure brain and induce Hsp70 protein. Brain Res 2008; 1234:183-97. [PMID: 18708034 DOI: 10.1016/j.brainres.2008.07.094] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 07/21/2008] [Accepted: 07/24/2008] [Indexed: 12/29/2022]
Abstract
This study examined very brief focal ischemia that simulates transient ischemic attacks (TIAs) that occur in humans. Adult rats were subjected to sham operations or 5 min, 10 min, or 2 h of middle cerebral artery (MCA) ischemia using the suture (thread) model. Hsp70 protein was induced 24 h, 48 h and 72 h later in neurons throughout the entire MCA territory in many but not all animals. Following 5- and 10-minute MCA occlusions, 9 of 32 animals (28%) had microinfarcts mostly in dorsal lateral striatum. Uncommon Hsp70 stained intracellular cytoplasmic inclusions, some of which co-localized with activated caspase-3, were detected in microglia, macrophages, astrocytes and oligodendrocytes. Hsp70 stained neurons were TUNEL negative at 24 h and 48 h whereas some Hsp70 stained neurons were TUNEL positive at 72 h after reperfusion. Hsp70 positive, activated "bushy" microglia and Hsp70 negative, activated "polarized" or rod-shaped microglia were located outside of the microinfarcts. Thus, experimental focal ischemia simulating TIAs can: induce Hsp70 protein throughout the ischemic vessel territory; produce Hsp70 protein positive glial inclusions; activate Hsp70 positive and negative microglia; and cause microinfarcts in some animals.
Collapse
Affiliation(s)
- Xinhua Zhan
- Department of Neurology and M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|