1
|
Zakharova IO, Bayunova LV, Avrova DK, Tretyakova AD, Shpakov AO, Avrova NF. The Autophagic and Apoptotic Death of Forebrain Neurons of Rats with Global Brain Ischemia Is Diminished by the Intranasal Administration of Insulin: Possible Mechanism of Its Action. Curr Issues Mol Biol 2024; 46:6580-6599. [PMID: 39057034 PMCID: PMC11276328 DOI: 10.3390/cimb46070392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Insulin is a promising neuroprotector. To better understand the mechanism of insulin action, it was important to show its ability to diminish autophagic neuronal death in animals with brain ischemic and reperfusion injury. In forebrain ischemia and reperfusion, the number of live neurons in the hippocampal CA1 region and frontal cortex of rats decreased to a large extent. Intracerebroventricular administration of the autophagy and apoptosis inhibitors to ischemic rats significantly increased the number of live neurons and showed that the main part of neurons died from autophagy and apoptosis. Intranasal administration of 0.5 IU of insulin per rat (before ischemia and daily during reperfusion) increased the number of live neurons in the hippocampal CA1 region and frontal brain cortex. In addition, insulin significantly diminished the level of autophagic marker LC3B-II in these forebrain regions, which markedly increased during ischemia and reperfusion. Our studies demonstrated for the first time the ability of insulin to decrease autophagic neuronal death, caused by brain ischemia and reperfusion. Insulin administered intranasally activated the Akt-kinase (activating the mTORC1 complex, which inhibits autophagy) and inhibited the AMP-activated protein kinase (which activates autophagy) in the hippocampus and frontal cortex of rats with brain ischemia and reperfusion.
Collapse
Affiliation(s)
| | | | | | | | | | - Natalia F. Avrova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez Av. 44, St. Petersburg 194223, Russia; (I.O.Z.); (L.V.B.); (D.K.A.); (A.D.T.); (A.O.S.)
| |
Collapse
|
2
|
Smith K, Fan J, Marriner GA, Gerdes J, Kessler R, Zinn KR. Distribution of insulin in primate brain following nose-to-brain transport. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12459. [PMID: 38469552 PMCID: PMC10925727 DOI: 10.1002/trc2.12459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/24/2024] [Accepted: 02/02/2024] [Indexed: 03/13/2024]
Abstract
Introduction Nose-to-brain (N2B) insulin delivery has potential for Alzheimer's disease (AD) therapy. However, clinical implementation has been challenging without methods to follow N2B delivery non-invasively. Positron emission tomography (PET) was applied to measure F-18-labeled insulin ([18F]FB-insulin) from intranasal dosing to brain uptake in non-human primates following N2B delivery. Methods [18F]FB-insulin was prepared by reacting A1,B29-di(tert-butyloxycarbonyl)insulin with [18F]-N-succinimidyl-4-fluorobenzoate. Three methods of N2B delivery for [18F]FB-insulin were compared - delivery as aerosol via tubing (rhesus macaque, n = 2), as aerosol via preplaced catheter (rhesus macaque, n = 3), and as solution via preplaced catheter (cynomolgus macaque, n = 3). Following dosing, dynamic PET imaging (120 min) quantified delivery efficiency to the nasal cavity and whole brain. Area under the time-activity curve was calculated for 46 regions of the cynomolgus macaque brain to determine regional [18F]FB-insulin levels. Results Liquid instillation of [18F]FB-insulin by catheter outperformed aerosol methods for delivery to the subject (39.89% injected dose vs 10.03% for aerosol via tubing, 0.17% for aerosol by catheter) and subsequently to brain (0.34% injected dose vs 0.00020% for aerosol via tubing, 0.05% for aerosol by catheter). [18F]FB-insulin was rapidly transferred across the cribriform plate to limbic and frontotemporal areas responsible for emotional and memory processing. [18F]FB-insulin half-life was longer in olfactory nerve projection sites with high insulin receptor density compared to the whole brain. Discussion The catheter-based liquid delivery approach combined with PET imaging successfully tracked the fate of N2B [18F]FB-insulin and is thought to be broadly applicable for assessments of other therapeutic agents. This method can be rapidly applied in humans to advance clinical evaluation of N2B insulin as an AD therapeutic. Highlights for [18F]FB-insulin passage across the cribriform plate was detected by PET.Intranasal [18F]FB-insulin reached the brain within 13 min.[18F]FB-insulin activity was highest in emotional and memory processing regions.Aerosol delivery was less efficient than liquid instillation by preplaced catheter.Insulin delivery to the cribriform plate was critical for arrival in the brain.
Collapse
Affiliation(s)
- Kylie Smith
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMichiganUSA
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
| | - Jinda Fan
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
- Department of ChemistryMichigan State UniversityEast LansingMichiganUSA
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
| | | | - John Gerdes
- Department of Biomedical and Pharmaceutical SciencesUniversity of MontanaMissoulaMontanaUSA
| | - Robert Kessler
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
| | - Kurt R. Zinn
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMichiganUSA
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
- Department of Small Animal Clinical SciencesMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
3
|
Talati CP, Lee JW, Lu S, Ojeda NB, Prakash V, Dankhara N, Nielson TC, Sandifer SP, Bidwell GL, Pang Y, Fan LW, Bhatt AJ. Intranasal insulin attenuates hypoxia-ischemia-induced short-term sensorimotor behavioral disturbances, neuronal apoptosis, and brain damage in neonatal rats. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 6:100123. [PMID: 38235171 PMCID: PMC10793091 DOI: 10.1016/j.crneur.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024] Open
Abstract
There is a significant need for additional therapy to improve outcomes for newborns with acute Hypoxic-ischemic (HI) encephalopathy (HIE). New evidence suggests that insulin could be neuroprotective. This study aimed to investigate whether intranasal insulin attenuates HI-induced brain damage and neurobehavioral dysfunction in neonatal rats. Postnatal day 10 (P10), Sprague-Dawley rat pups were randomly divided into Sham + Vehicle, Sham + Insulin, HI + Vehicle, and HI + Insulin groups with equal male-to-female ratios. Pups either had HI by permanent ligation of the right common carotid artery followed by 90 min of hypoxia (8% O2) or sham surgery followed by room air exposure. Immediately after HI or Sham, pups were given fluorescence-tagged insulin (Alex-546-insulin)/vehicle, human insulin (25 μg), or vehicle in each nare under anesthesia. Shortly after administration, widespread Alex-546-insulin-binding cells were detected in the brain, primarily co-localized with neuronal nuclei-positive neurons on double-immunostaining. In the hippocampus, phospho-Akt was activated in a subset of Alex-546-insulin double-labeled cells, suggesting activation of the Akt/PI3K pathway in these neurons. Intranasal insulin (InInsulin) reduced HI-induced sensorimotor behavioral disturbances at P11. InInsulin prevented HI-induced increased Fluoro-Jade C+ degenerated neurons, cleaved caspase 3+ neurons, and volume loss in the ipsilateral brain at P11. There was no sex-specific response to HI or insulin. The findings confirm that intranasal insulin provides neuroprotection against HI brain injury in P10 rats associated with activation of intracellular cell survival signaling. If further pre-clinical research shows long-term benefits, intranasal insulin has the potential to be a promising non-invasive therapy to improve outcomes for newborns with HIE.
Collapse
Affiliation(s)
- Chirag P. Talati
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jonathan W. Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Silu Lu
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Norma B. Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Varsha Prakash
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Nilesh Dankhara
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Tanner C. Nielson
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Sara P. Sandifer
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Abhay J. Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| |
Collapse
|
4
|
Zorina II, Avrova NF, Zakharova IO, Shpakov AO. Prospects for the Use of Intranasally Administered Insulin and Insulin-Like Growth Factor-1 in Cerebral Ischemia. BIOCHEMISTRY (MOSCOW) 2023; 88:374-391. [PMID: 37076284 DOI: 10.1134/s0006297923030070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
Abstract
Current approaches to the treatment of stroke have significant limitations, and neuroprotective therapy is ineffective. In view of this, searching for effective neuroprotectors and developing new neuroprotective strategies remain a pressing topic in research of cerebral ischemia. Insulin and insulin-like growth factor-1 (IGF-1) play a key role in the brain functioning by regulating the growth, differentiation, and survival of neurons, neuronal plasticity, food intake, peripheral metabolism, and endocrine functions. Insulin and IGF-1 produce multiple effects in the brain, including neuroprotective action in cerebral ischemia and stroke. Experiments in animals and cell cultures have shown that under hypoxic conditions, insulin and IGF-1 improve energy metabolism in neurons and glial cells, promote blood microcirculation in the brain, restore nerve cell functions and neurotransmission, and produce the anti-inflammatory and antiapoptotic effects on brain cells. The intranasal route of insulin and IGF-1 administration is of particular interest in the clinical practice, since it allows controlled delivery of these hormones directly to the brain, bypassing the blood-brain barrier. Intranasally administered insulin alleviated cognitive impairments in elderly people with neurodegenerative and metabolic disorders; intranasally administered insulin and IGF-1 promoted survival of animals with ischemic stroke. The review discusses the published data and results of our own studies on the mechanisms of neuroprotective action of intranasally administered insulin and IGF-1 in cerebral ischemia, as well as the prospects of using these hormones for normalization of CNS functions and reduction of neurodegenerative changes in this pathology.
Collapse
Affiliation(s)
- Inna I Zorina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint-Petersburg, 194223, Russia.
| | - Natalia F Avrova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint-Petersburg, 194223, Russia
| | - Irina O Zakharova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint-Petersburg, 194223, Russia
| | - Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint-Petersburg, 194223, Russia
| |
Collapse
|
5
|
Shpakov AO, Zorina II, Derkach KV. Hot Spots for the Use of Intranasal Insulin: Cerebral Ischemia, Brain Injury, Diabetes Mellitus, Endocrine Disorders and Postoperative Delirium. Int J Mol Sci 2023; 24:3278. [PMID: 36834685 PMCID: PMC9962062 DOI: 10.3390/ijms24043278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A decrease in the activity of the insulin signaling system of the brain, due to both central insulin resistance and insulin deficiency, leads to neurodegeneration and impaired regulation of appetite, metabolism, endocrine functions. This is due to the neuroprotective properties of brain insulin and its leading role in maintaining glucose homeostasis in the brain, as well as in the regulation of the brain signaling network responsible for the functioning of the nervous, endocrine, and other systems. One of the approaches to restore the activity of the insulin system of the brain is the use of intranasally administered insulin (INI). Currently, INI is being considered as a promising drug to treat Alzheimer's disease and mild cognitive impairment. The clinical application of INI is being developed for the treatment of other neurodegenerative diseases and improve cognitive abilities in stress, overwork, and depression. At the same time, much attention has recently been paid to the prospects of using INI for the treatment of cerebral ischemia, traumatic brain injuries, and postoperative delirium (after anesthesia), as well as diabetes mellitus and its complications, including dysfunctions in the gonadal and thyroid axes. This review is devoted to the prospects and current trends in the use of INI for the treatment of these diseases, which, although differing in etiology and pathogenesis, are characterized by impaired insulin signaling in the brain.
Collapse
Affiliation(s)
- Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | | | | |
Collapse
|
6
|
Deng A, Ma L, Ji Q, Xing J, Qin J, Zhou X, Wang X, Wang S, Wu J, Chen X. Activation of the Akt/FoxO3 signaling pathway enhances oxidative stress-induced autophagy and alleviates brain damage in a rat model of ischemic stroke. Can J Physiol Pharmacol 2023; 101:18-26. [PMID: 36315971 DOI: 10.1139/cjpp-2022-0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autophagy has been implicated in stroke. Our previous study showed that the FoxO3 transcription factor promotes autophagy after transient cerebral ischemia/reperfusion (I/R). However, whether the Akt/FoxO3 signaling pathway plays a regulatory role in autophagy in cerebral I/R-induced oxidative stress injury is still unclear. The present study aims to investigate the effects of the Akt/FoxO3 signaling pathway on autophagy activation and neuronal injury in vitro and in vivo. By employing LY294002 or insulin to regulate the Akt/FoxO3 signaling pathway, we found that insulin pretreatment increased cell viability, decreased reactive oxygen species production, and enhanced the expression of antiapoptotic and autophagy-related proteins following H2O2 injury in HT22 cells. In addition, insulin significantly decreased neurological deficit scores and infarct volume and increased the expression of antiapoptotic and autophagy-related proteins following I/R injury in rats. However, LY294002 showed the opposite effects under these conditions. Altogether, these results indicate that Akt/FoxO3 signaling pathway activation inhibited oxidative stress-mediated cell death through activation of autophagy. Our study supports a critical role for the Akt/FoxO3 signaling pathway in autophagy activation in stroke.
Collapse
Affiliation(s)
- Aiqing Deng
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, People's Republic of China
| | - Limin Ma
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Qiuhong Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, People's Republic of China
| | - Jiajun Xing
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Jianxin Qin
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Xueli Zhou
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Xin Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Shouyan Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Jianjun Wu
- Department of Hepatobiliary, Nantong Third Hospital Affiliated to Nantong University, Nantong 226006, People's Republic of China
| | - Xia Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| |
Collapse
|
7
|
Yao M, Hao Y, Wang T, Xie M, Li H, Feng J, Feng L, Ma D. A review of stress-induced hyperglycaemia in the context of acute ischaemic stroke: Definition, underlying mechanisms, and the status of insulin therapy. Front Neurol 2023; 14:1149671. [PMID: 37025208 PMCID: PMC10070880 DOI: 10.3389/fneur.2023.1149671] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 02/21/2023] [Indexed: 04/08/2023] Open
Abstract
The transient elevation of blood glucose produced following acute ischaemic stroke (AIS) has been described as stress-induced hyperglycaemia (SIH). SIH is common even in patients with AIS who have no previous diagnosis of diabetes mellitus. Elevated blood glucose levels during admission and hospitalization are strongly associated with enlarged infarct size and adverse prognosis in AIS patients. However, insulin-intensive glucose control therapy defined by admission blood glucose for SIH has not achieved the desired results, and new treatment ideas are urgently required. First, we explore the various definitions of SIH in the context of AIS and their predictive value in adverse outcomes. Then, we briefly discuss the mechanisms by which SIH arises, describing the dual effects of elevated glucose levels on the central nervous system. Finally, although preclinical studies support lowering blood glucose levels using insulin, the clinical outcomes of intensive glucose control are not promising. We discuss the reasons for this phenomenon.
Collapse
Affiliation(s)
- Mengyue Yao
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tian Wang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Meizhen Xie
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hui Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Liangshu Feng
- Stroke Centre, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
- Liangshu Feng
| | - Di Ma
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Di Ma
| |
Collapse
|
8
|
Zakharova IO, Bayunova LV, Zorina II, Shpakov AO, Avrova NF. Insulin and Brain Gangliosides Prevent Metabolic Disorders Caused by Activation of Free Radical Reactions after Two-Vessel Ischemia–Reperfusion Injury to the Rat Forebrain. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022010240] [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]
|
9
|
Mejido DC, Peny JA, Vieira MN, Ferreira ST, De Felice FG. Insulin and leptin as potential cognitive enhancers in metabolic disorders and Alzheimer's disease. Neuropharmacology 2020; 171:108115. [DOI: 10.1016/j.neuropharm.2020.108115] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 02/08/2023]
|
10
|
Cytochrome c phosphorylation: Control of mitochondrial electron transport chain flux and apoptosis. Int J Biochem Cell Biol 2020; 121:105704. [PMID: 32023432 DOI: 10.1016/j.biocel.2020.105704] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/30/2022]
Abstract
Cytochrome c (Cytc)1is a cellular life and death decision molecule that regulates cellular energy supply and apoptosis through tissue specific post-translational modifications. Cytc is an electron carrier in the mitochondrial electron transport chain (ETC) and thus central for aerobic energy production. Under conditions of cellular stress, Cytc release from the mitochondria is a committing step for apoptosis, leading to apoptosome formation, caspase activation, and cell death. Recently, Cytc was shown to be a target of cellular signaling pathways that regulate the functions of Cytc by tissue-specific phosphorylations. So far five phosphorylation sites of Cytc have been mapped and functionally characterized, Tyr97, Tyr48, Thr28, Ser47, and Thr58. All five phosphorylations partially inhibit respiration, which we propose results in optimal intermediate mitochondrial membrane potentials and low ROS production under normal conditions. Four of the phosphorylations result in inhibition of the apoptotic functions of Cytc, suggesting a cytoprotective role for phosphorylated Cytc. Interestingly, these phosphorylations are lost during stress conditions such as ischemia. This results in maximal ETC flux during reperfusion, mitochondrial membrane potential hyperpolarization, excessive ROS generation, and apoptosis. We here present a new model proposing that the electron transfer from Cytc to cytochrome c oxidase is the rate-limiting step of the ETC, which is regulated via post-translational modifications of Cytc. This regulation may be dysfunctional in disease conditions such as ischemia-reperfusion injury and neurodegenerative disorders through increased ROS, or cancer, where post-translational modifications on Cytc may provide a mechanism to evade apoptosis.
Collapse
|
11
|
Hiramoto JS, Hoffman M, Gasper W, Reilly L, Chuter T. Strict Control of Blood Glucose With an Intravenous Insulin Infusion Decreases the Risk of Post-operative Lower Extremity Weakness After Complex Endovascular Aortic Aneurysm Repair. Eur J Vasc Endovasc Surg 2019; 58:848-853. [DOI: 10.1016/j.ejvs.2019.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/03/2019] [Accepted: 03/16/2019] [Indexed: 12/31/2022]
|
12
|
Russo V, Candeloro P, Malara N, Perozziello G, Iannone M, Scicchitano M, Mollace R, Musolino V, Gliozzi M, Carresi C, Morittu VM, Gratteri S, Palma E, Muscoli C, Di Fabrizio E, Mollace V. Key Role of Cytochrome C for Apoptosis Detection Using Raman Microimaging in an Animal Model of Brain Ischemia with Insulin Treatment. APPLIED SPECTROSCOPY 2019; 73:1208-1217. [PMID: 31219322 DOI: 10.1177/0003702819858671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Brain ischemia represents a leading cause of death and disability in industrialized countries. To date, therapeutic intervention is largely unsatisfactory and novel strategies are required for getting better protection of neurons injured by cerebral blood flow restriction. Recent evidence suggests that brain insulin leads to protection of neuronal population undergoing apoptotic cell death via modulation of oxidative stress and mitochondrial cytochrome c (CytC), an effect to be better clarified. In this work, we investigate on the effect of insulin given intracerebroventricular (ICV) before inducing a transient global ischemia by bilateral occlusion of the common carotid arteries (BCCO) in Mongolian gerbils (MG). The transient (3 min) global ischemia in MG is observed to produce neurodegenerative effect mainly into CA3 hippocampal region, 72 h after cerebral blood restriction. Intracerebroventricular microinfusion of insulin significantly prevents the apoptosis of CA3 hippocampal neurons. Histological observation, after hematoxylin and eosin staining, puts in evidence the neuroprotective role of insulin, but Raman microimaging provides a clearer insight in the CytC mechanism underlying the apoptotic process. Above all, CytC has been revealed to be an outstanding, innate Raman marker for monitoring the cells status, thanks to its resonant scattering at 530 nm of incident wavelength and to its crucial role in the early stages of cells apoptosis. These data support the hypothesis of an insulin-dependent neuroprotection and antiapoptotic mechanism occurring in the brain of MG undergoing transient brain ischemia. The observed effects occurred without any peripheral change on serum glucose levels, suggesting an alternative mechanism of insulin-induced neuroprotection.
Collapse
Affiliation(s)
- Vanessa Russo
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Association: Exchanger-Share Your Science, Complesso "Nini Barbieri," Catanzaro, Italy
| | - Patrizio Candeloro
- BioNEM Laboratory, Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Natalia Malara
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- BioNEM Laboratory, Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Gerardo Perozziello
- BioNEM Laboratory, Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Michelangelo Iannone
- CNR, Neuroscience Institute, Pharmacology Section, Complesso "Nini Barbieri," Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
| | - Rocco Mollace
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
| | - Micaela Gliozzi
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
| | - Cristina Carresi
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
| | - Valeria M Morittu
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
| | - Santo Gratteri
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
| | - Ernesto Palma
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
| | - Carolina Muscoli
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
- Centro del farmaco (IRCCS), Rome, Italy
| | - Enzo Di Fabrizio
- BioNEM Laboratory, Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, Italy
- KAUST (King Abdullah University of Science and Technology), PSE and BESE Divisions, Thuwal, Kingdom of Saudi Arabia
| | - Vincenzo Mollace
- IRC-FSH Interregional Center for Food Safety and Health, University "Magna Graecia" of Catanzaro, Italy
- Nutramed S.C.A.R.L., Complesso "Nini Barbieri", Roccelletta di Borgia, Catanzaro, Italy 88100
- Centro del farmaco (IRCCS), Rome, Italy
| |
Collapse
|
13
|
Kim B, Elzinga SE, Henn RE, McGinley LM, Feldman EL. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer's disease. Neurobiol Dis 2019; 132:104541. [PMID: 31349033 DOI: 10.1016/j.nbd.2019.104541] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a growing problem worldwide, and there are currently no effective treatments for this devastating disease. The neurotrophic growth factors insulin and insulin-like growth factor-I (IGF-I) are currently being investigated as potential therapeutic approaches for AD in preclinical and clinical studies. However, given that the metabolic syndrome (MetS) and diabetes are risk factors for AD, it is unknown how associated insulin resistance (IR) in the brain may impact the effectiveness of these therapies for AD. In this report, we therefore investigated the mechanisms underlying the effects of insulin and IGF-I on AD-associated pathology in the context of IR, with particular emphasis on phosphorylation of amyloid precursor protein (APP), a key step in promoting amyloid plaque formation in AD. Both insulin and IGF-I decreased APP phosphorylation in cultured primary cortical neurons, supporting their therapeutic use in AD. Induction of IR blocked the beneficial effect of insulin and reduced the effect of IGF-I on APP dephosphorylation. These effects were mediated by the phosphatidylinositol 3-kinase (PI3-K)/protein kinase B (Akt) pathway, as inhibition of this pathway during IR restored the effect of IGF-I on APP dephosphorylation. Finally, we explored the translational relevance of these results in vivo by demonstrating that high fat diet fed mice, a robust model of IR and MetS, exhibited the expected increased brain APP phosphorylation. Overall, these data suggest that the beneficial therapeutic effect of insulin and IGF-I on APP phosphorylation is negatively impacted by IR, and suggest that insulin and IGF-I alone may not be appropriate therapies for AD patients with IR, MetS, or diabetes.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Sarah E Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Rosemary E Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America.
| |
Collapse
|
14
|
Mormile R. Neonates of diabetic mothers: The starting point for developing novel therapeutic approaches to ischemic heart and brain? Med Hypotheses 2016; 96:75-77. [DOI: 10.1016/j.mehy.2016.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/16/2016] [Indexed: 10/20/2022]
|
15
|
Nagalski A, Kozinski K, Wisniewska MB. Metabolic pathways in the periphery and brain: Contribution to mental disorders? Int J Biochem Cell Biol 2016; 80:19-30. [PMID: 27644152 DOI: 10.1016/j.biocel.2016.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/20/2022]
Abstract
The association between mental disorders and diabetes has a long history. Recent large-scale, well-controlled epidemiological studies confirmed a link between diabetes and psychiatric illnesses. The scope of this review is to summarize our current understanding of this relationship from a molecular perspective. We first discuss the potential contribution of diabetes-associated metabolic impairments to the etiology of mental conditions. Then, we focus on possible shared molecular risk factors and mechanisms. Simple comorbidity, shared susceptibility loci, and common pathophysiological processes in diabetes and mental illnesses have changed our traditional way of thinking about mental illness. We conclude that schizophrenia and affective disorders are not limited to an imbalance in dopaminergic and serotoninergic neurotransmission in the brain. They are also systemic disorders that can be considered, to some extent, as metabolic disorders.
Collapse
Affiliation(s)
- Andrzej Nagalski
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Kamil Kozinski
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Marta B Wisniewska
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland.
| |
Collapse
|
16
|
Zhang H, Liu Y, Guan S, Qu D, Wang L, Wang X, Li X, Zhou S, Zhou Y, Wang N, Meng J, Ma X. An Orally Active Allosteric GLP-1 Receptor Agonist Is Neuroprotective in Cellular and Rodent Models of Stroke. PLoS One 2016; 11:e0148827. [PMID: 26863436 PMCID: PMC4749391 DOI: 10.1371/journal.pone.0148827] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/21/2016] [Indexed: 12/25/2022] Open
Abstract
Diabetes is a major risk factor for the development of stroke. Glucagon-like peptide-1 receptor (GLP-1R) agonists have been in clinical use for the treatment of diabetes and also been reported to be neuroprotective in ischemic stroke. The quinoxaline 6,7-dichloro-2-methylsulfonyl-3-N-tert- butylaminoquinoxaline (DMB) is an agonist and allosteric modulator of the GLP-1R with the potential to increase the affinity of GLP-1 for its receptor. The aim of this study was to evaluate the neuroprotective effects of DMB on transient focal cerebral ischemia. In cultured cortical neurons, DMB activated the GLP-1R, leading to increased intracellular cAMP levels with an EC50 value about 100 fold that of exendin-4. Pretreatment of neurons with DMB protected against necrotic and apoptotic cell death was induced by oxygen-glucose deprivation (OGD). The neuroprotective effects of DMB were blocked by GLP-1R knockdown with shRNA but not by GLP-1R antagonism. In C57BL/6 mice, DMB was orally administered 30 min prior to middle cerebral artery occlusion (MCAO) surgery. DMB markedly reduced the cerebral infarct size and neurological deficits caused by MCAO and reperfusion. The neuroprotective effects were mediated by activation of the GLP-1R through the cAMP-PKA-CREB signaling pathway. DMB exhibited anti-apoptotic effects by modulating Bcl-2 family members. These results provide evidence that DMB, a small molecular GLP-1R agonist, attenuates transient focal cerebral ischemia injury and inhibits neuronal apoptosis induced by MCAO. Taken together, these data suggest that DMB is a potential neuroprotective agent against cerebral ischemia.
Collapse
Affiliation(s)
- Huinan Zhang
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Yunhan Liu
- School of Nurse, the Fourth Military Medical University, Xi’an, China
| | - Shaoyu Guan
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Di Qu
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Ling Wang
- Department of Health Statistics, Faculty of Preventative Medicine, the Fourth Military Medical University, Xi’an, China
| | - Xinshang Wang
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Xubo Li
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Shimeng Zhou
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Ying Zhou
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Ning Wang
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
| | - Jingru Meng
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
- * E-mail: . (XM); (JM)
| | - Xue Ma
- Department of Pharmacology, School of Pharmacy, the Fourth Military Medical University, Xi’an, China
- * E-mail: . (XM); (JM)
| |
Collapse
|
17
|
Abstract
Objective: To analyze the mechanism of neuroprotection of insulin and which blood glucose range was benefit for insulin exerting neuroprotective action. Data Sources: The study is based on the data from PubMed. Study Selection: Articles were selected with the search terms “insulin”, “blood glucose”, “neuroprotection”, “brain”, “glycogen”, “cerebral ischemia”, “neuronal necrosis”, “glutamate”, “γ-aminobutyric acid”. Results: Insulin has neuroprotection. The mechanisms include the regulation of neurotransmitter, promoting glycogen synthesis, and inhibition of neuronal necrosis and apoptosis. Insulin could play its role in neuroprotection by avoiding hypoglycemia and hyperglycemia. Conclusions: Intermittent and long-term infusion insulin may be a benefit for patients with ischemic brain damage at blood glucose 6–9 mmol/L.
Collapse
Affiliation(s)
| | - Yu Pei
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
18
|
Zhang H, Meng J, Li X, Zhou S, Qu D, Wang N, Jia M, Ma X, Luo X. Pro-GLP-1, a Pro-drug of GLP-1, is neuroprotective in cerebral ischemia. Eur J Pharm Sci 2015; 70:82-91. [DOI: 10.1016/j.ejps.2015.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 01/18/2023]
|
19
|
Pilchova I, Klacanova K, Chomova M, Tatarkova Z, Dobrota D, Racay P. Possible contribution of proteins of Bcl-2 family in neuronal death following transient global brain ischemia. Cell Mol Neurobiol 2015; 35:23-31. [PMID: 25187358 DOI: 10.1007/s10571-014-0104-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 08/23/2014] [Indexed: 01/08/2023]
Abstract
Proteins of Bcl-2 family are crucial regulators of intrinsic (mitochondrial) pathway of apoptosis that is implicated among the mechanisms of ischemic neuronal death. Initiation of mitochondrial apoptosis depends on changes of equilibrium between anti-apoptotic and pro-apoptotic proteins of Bcl-2 family as well as on translocation of pro-apoptotic proteins of Bcl-2 family to mitochondria. The aim of this work was to study the effect of transient global brain ischemia on expression and intracellular distribution of proteins of Bcl-2 family in relation to the ischemia-induced changes of ERK and Akt kinase pathways as well as disturbances in ubiquitin proteasome system. Using four vessel occlusion model of transient global brain ischemia, we have shown that both ischemia in duration of 15 min and the same ischemia followed by 1, 3, 24, and 72 h of reperfusion did not affect the levels of either pro-apoptotic (Bad, PUMA, Bim, Bax, Noxa) or anti-apoptotic (Bcl-2, Bcl-xl, Mcl-1) proteins of Bcl-2 family in total cell extracts from rat hippocampus. However, significantly elevated level of Bad protein in the mitochondria isolated from rat hippocampus was observed already 1 h after ischemia and remained elevated 3 and 24 h after ischemia. We did not observe significant changes of the levels of Puma, Bax, Bcl-2, and Bcl-xl in the mitochondria after ischemia and ischemia followed by reperfusion. Our results might indicate possible involvement of Bad translocation to mitochondria in the mechanisms of neuronal death following transient global brain ischemia.
Collapse
Affiliation(s)
- Ivana Pilchova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | | | | | | | | | | |
Collapse
|
20
|
Herson PS, Traystman RJ. Animal models of stroke: translational potential at present and in 2050. FUTURE NEUROLOGY 2014; 9:541-551. [PMID: 25530721 DOI: 10.2217/fnl.14.44] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Translation from basic science bench research in ischemic stroke to bedside treatment of patients suffering ischemic stroke remains a difficult challenge. Despite literally hundreds of compounds and interventions that provide benefit in experimental models of cerebral ischemia, efficacy in humans remains to be demonstrated. The reasons for failure to translate the extensive positive basic science findings to successful clinical trials have been the focus of discussion for years. Some attribute the failure to flaws in clinical trial design, others question the predictive value of current animal models and some question the quality of preclinical data. It is likely that a combination of all these shortcomings have ultimately led to the failure. The purpose of this review is to analyze the commonly used animal models used in the field today, provide a framework for understanding the current state of basic science research in the ischemic stroke field and discuss a path forward.
Collapse
Affiliation(s)
- Paco S Herson
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, 12800 E 19th Avenue, Aurora, CO 80045, USA ; Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, 12800 E 19th Avenue, Aurora, CO 80045, USA
| | - Richard J Traystman
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, 12800 E 19th Avenue, Aurora, CO 80045, USA ; Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, 12800 E 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Linying Z, Wei W, Minxia W, Wenmin Z, Liangcheng Z. Neuroprotective effects of neuregulin-1 ß on oligodendrocyte type 2 astrocyte progenitors following oxygen and glucose deprivation. Pediatr Neurol 2014; 50:357-62. [PMID: 24529326 DOI: 10.1016/j.pediatrneurol.2013.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 11/27/2013] [Accepted: 12/07/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hypoxic-ischemic brain injury in neonates, especially in premature infants, is one of the main contributors to the mortality of newborns and can cause nervous system dysfunction in children. The major pathogenesis seems to be cerebral ischemia/reperfusion in the immature white matter that preferentially targets vulnerable premyelinating oligodendrocytes. OBJECTIVES The goal of this study was to culture oligodendrocyte type 2 astrocyte cells in an oxygen and glucose deprivation environment to simulate ischemia injury and examine the cellular and molecular mechanisms involved in the neuroprotective effects of neuregulin-1ß on ischemia-induced immature oligodendrocytes. METHODS Oligodendrocyte type 2 astrocyte cells were cultured from neonatal Sprague-Dawley rat cerebra. The cells were divided into two groups: one was subjected to oxygen and glucose deprivation for 9 hours and the other was treated with 50 ng/mL or 100 ng/mL neuregulin-1β during oxygen and glucose deprivation. Cell survival was determined by Trypan Blue staining and cell apoptosis were observed by fluorescein isothiocyanate-Annexin V and propidium iodide double staining. To study if the PI3K-Akt signaling pathway was involved in the mechanism of protective effect of neuregulin-1ß, Western blot analysis was used to quantitative the changes of protein. RESULTS Treatment with neuregulin-1ß within the period of oxygen and glucose deprivation significantly increased cell survival and also resulted in a significant decrease in cell apoptosis. The neuroprotective effects of neuregulin-1ß were prevented by treatment with Ly294002, an inhibitor of the phosphatidylinositol-3-kinase/Akt pathway. CONCLUSIONS These results suggest that neuregulin-1ß could protect the oligodendrocyte type 2 astrocyte progenitors against hypoxic injury, and the mechanism may be associated with the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Zhou Linying
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Centre of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Wang Wei
- Centre of Neuroscience, Fujian Medical University, Fuzhou, China.
| | - Wu Minxia
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zhang Wenmin
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zhang Liangcheng
- Department of Anaesthesiology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
22
|
Zhao S, Fu J, Liu F, Rastogi R, Zhang J, Zhao Y. Small interfering RNA directed against CTMP reduces acute traumatic brain injury in a mouse model by activating Akt. Neurol Res 2014; 36:483-90. [PMID: 24670215 DOI: 10.1179/1743132814y.0000000353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Protein kinase B (PKB/Akt), which is phosphorylated and activated by upstream activators, exerts critical neuroprotective effects by phosphorylating downstream targets after traumatic brain injury (TBI). Studies on the regulation of Akt will be crucial for our understanding of neuronal survival. The goal of this study is to investigate the effects of carboxyl-terminal modulator protein (CTMP) on phosphorylation of Akt and neurological function in a mouse model of TBI. METHODS Traumatic brain injury in mice was performed by a controlled cortical impact device. The expression of Akt, phospho-Akt, and CTMP was examined in the injured cortices by immunohistochemistry and Western blot analysis. To determine the effects of CTMP, small interfering RNAs (siRNAs) directed against CTMP were injected in mice with TBI, and the expression of phosphorylated Akt and neurological function were evaluated. RESULTS Phospho-Akt significantly increased at 4 hours post-TBI in the nucleus (P < 0.01) and remained at high levels until 72 hours after TBI, as shown by Western blot analysis. In the cytosol, the expression of phospho-Akt reached its peak at 4 hours post-TBI, but decreased markedly at 24 hours and maintained below pre-TBI levels until 72 hours post-TBI. Interestingly, the expression of CTMP significantly increased 4 hours after TBI (P < 0.01) and sustained those levels until 72 hours without dramatic changes. Treatment with CTMP siRNA effectively augmented the phosphorylation of Akt and significantly improved the neurological functional recovery up to 28 days post-TBI. CONCLUSION We conclude that Akt is phosphorylated and translocated to nucleus after TBI to exert neuroprotective effects. However, CTMP is simultaneously triggered to inhibit the phosphorylation of Akt. Inhibition of CTMP by siRNA improves the recovery of neurological functions after TBI.
Collapse
|
23
|
Zhang X, Yan X, Gorman J, Hoffman SN, Zhang L, Boscarino JA. Perioperative hyperglycemia is associated with postoperative neurocognitive disorders after cardiac surgery. Neuropsychiatr Dis Treat 2014; 10:361-70. [PMID: 24570589 PMCID: PMC3933727 DOI: 10.2147/ndt.s57761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Neurocognitive disorders commonly occur following cardiac surgery. However, the underlying etiology of these disorders is not well understood. The current study examined the association between perioperative glucose levels and other risk factors and the onset of neurocognitive disorders in adult patients following coronary artery bypass and/or valvular surgery. METHODS Adult patients who underwent their first cardiac surgery at a large tertiary care medical center were identified and those with neurocognitive disorders prior to surgery were excluded. Demographic, perioperative, and postoperative neurocognitive outcome data were extracted from the Society for Thoracic Surgery database, and from electronic medical records, between January 2004 and June 2009. Multiple clinical risk factors and measures associated with insulin resistance, such as hyperglycemia, were assessed. Multivariable Cox competing risk survival models were used to assess hyperglycemia and postoperative neurocognitive disorders at follow up, adjusting for other risk factors and confounding variables. RESULTS Of the 855 patients included in the study, 271 (31.7%) had new onset neurocognitive disorders at follow-up. Age, sex, New York Heart Failure (NYHF) Class, length of postoperative intensive care unit stay, perioperative blood product transfusion, and other key factors were identified and assessed as potential risk factors (or confounders) for neurocognitive disorders at follow-up. Bivariate analyses suggested that new onset neurocognitive disorders were associated with NYHF Class, cardiopulmonary bypass, history of diabetes, intraoperative blood product use, and number of diseased coronary vessels, which are commonly-accepted risk factors in cardiac surgery. In addition, higher first glucose level (median =116 mg/dL) and higher peak glucose >169 mg/dL were identified as risk factors. Male sex and nonuse of intra-operative blood products appeared to be protective. Controlling for potential risk factors and confounders, multivariable Cox survival models suggested that increased perioperative first glucose measured in 20 unit increments, was significantly associated with the onset of postoperative neurocognitive disorders at follow-up (hazard ratio [HR] =1.16, P<0.001) and that women had an elevated risk for this outcome (HR =4.18, P=0.01). CONCLUSION Our study suggests that perioperative hyperglycemia was associated with new onset of postoperative neurocognitive disorders in adult patients after cardiac surgery, and that men tended to be protected from these outcomes. These findings may suggest a need for the revision of clinical protocols for perioperative insulin therapy to prevent long-term neurocognitive complications.
Collapse
Affiliation(s)
- Xiaopeng Zhang
- Department of Anesthesiology, Geisinger Medical Center, Danville, PA, USA
| | - Xiaowei Yan
- Center for Health Research, Geisinger Clinic, Danville, PA, USA
| | - Jennifer Gorman
- Center for Health Research, Geisinger Clinic, Danville, PA, USA
| | - Stuart N Hoffman
- Department of Neurology, Geisinger Medical Center, Danville, PA, USA
| | - Li Zhang
- Department of Anesthesiology, Geisinger Medical Center, Danville, PA, USA
| | | |
Collapse
|
24
|
Ouyang YB, Giffard RG. MicroRNAs affect BCL-2 family proteins in the setting of cerebral ischemia. Neurochem Int 2013; 77:2-8. [PMID: 24373752 DOI: 10.1016/j.neuint.2013.12.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/09/2013] [Accepted: 12/16/2013] [Indexed: 02/01/2023]
Abstract
The BCL-2 family is centrally involved in the mechanism of cell death after cerebral ischemia. It is well known that the proteins of the BCL-2 family are key regulators of apoptosis through controlling mitochondrial outer membrane permeabilization. Recent findings suggest that many BCL-2 family members are also directly involved in controlling transmission of Ca(2+) from the endoplasmic reticulum (ER) to mitochondria through a specialization called the mitochondria-associated ER membrane (MAM). Increasing evidence supports the involvement of microRNAs (miRNAs), some of them targeting BCL-2 family proteins, in the regulation of cerebral ischemia. In this mini-review, after highlighting current knowledge about the multiple functions of BCL-2 family proteins and summarizing their relationship to outcome from cerebral ischemia, we focus on the regulation of BCL-2 family proteins by miRNAs, especially miR-29 which targets multiple BCL-2 family proteins.
Collapse
Affiliation(s)
- Yi-Bing Ouyang
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Rona G Giffard
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Sanderson TH, Mahapatra G, Pecina P, Ji Q, Yu K, Sinkler C, Varughese A, Kumar R, Bukowski MJ, Tousignant RN, Salomon AR, Lee I, Hüttemann M. Cytochrome C is tyrosine 97 phosphorylated by neuroprotective insulin treatment. PLoS One 2013; 8:e78627. [PMID: 24223835 PMCID: PMC3818486 DOI: 10.1371/journal.pone.0078627] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/13/2013] [Indexed: 12/23/2022] Open
Abstract
Recent advancements in isolation techniques for cytochrome c (Cytc) have allowed us to discover post-translational modifications of this protein. We previously identified two distinct tyrosine phosphorylated residues on Cytc in mammalian liver and heart that alter its electron transfer kinetics and the ability to induce apoptosis. Here we investigated the phosphorylation status of Cytc in ischemic brain and sought to determine if insulin-induced neuroprotection and inhibition of Cytc release was associated with phosphorylation of Cytc. Using an animal model of global brain ischemia, we found a ∼50% decrease in neuronal death in the CA1 hippocampal region with post-ischemic insulin administration. This insulin-mediated increase in neuronal survival was associated with inhibition of Cytc release at 24 hours of reperfusion. To investigate possible changes in the phosphorylation state of Cytc we first isolated the protein from ischemic pig brain and brain that was treated with insulin. Ischemic brains demonstrated no detectable tyrosine phosphorylation. In contrast Cytc isolated from brains treated with insulin showed robust phosphorylation of Cytc, and the phosphorylation site was unambiguously identified as Tyr97 by immobilized metal affinity chromatography/nano-liquid chromatography/electrospray ionization mass spectrometry. We next confirmed these results in rats by in vivo application of insulin in the absence or presence of global brain ischemia and determined that Cytc Tyr97-phosphorylation is strongly induced under both conditions but cannot be detected in untreated controls. These data suggest a mechanism whereby Cytc is targeted for phosphorylation by insulin signaling, which may prevent its release from the mitochondria and the induction of apoptosis.
Collapse
Affiliation(s)
- Thomas H. Sanderson
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Gargi Mahapatra
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Petr Pecina
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Institute of Physiology and Center for Applied Genomics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Qinqin Ji
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Kebing Yu
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Christopher Sinkler
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Ashwathy Varughese
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Rita Kumar
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Melissa J. Bukowski
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Renee N. Tousignant
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Arthur R. Salomon
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Maik Hüttemann
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
26
|
Sanderson TH, Wider JM. 2-vessel occlusion/hypotension: a rat model of global brain ischemia. J Vis Exp 2013. [PMID: 23851591 DOI: 10.3791/50173] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cardiac arrest followed by resuscitation often results in dramatic brain damage caused by ischemia and subsequent reperfusion of the brain. Global brain ischemia produces damage to specific brain regions shown to be highly sensitive to ischemia (1). Hippocampal neurons have higher sensitivity to ischemic insults compared to other cell populations, and specifically, the CA1 region of the hippocampus is particularly vulnerable to ischemia/reperfusion (2). The design of therapeutic interventions, or study of mechanisms involved in cerebral damage, requires a model that produces damage similar to the clinical condition and in a reproducible manner. Bilateral carotid vessel occlusion with hypotension (2VOH) is a model that produces reversible forebrain ischemia, emulating the cerebral events that can occur during cardiac arrest and resuscitation. We describe a model modified from Smith et al. (1984) (2), as first presented in its current form in Sanderson, et al. (2008) (3), which produces reproducible injury to selectively vulnerable brain regions (3-6). The reliability of this model is dictated by precise control of systemic blood pressure during applied hypotension, the duration of ischemia, close temperature control, a specific anesthesia regimen, and diligent post-operative care. An 8-minute ischemic insult produces cell death of CA1 hippocampal neurons that progresses over the course of 6 to 24 hr of reperfusion, while less vulnerable brain regions are spared. This progressive cell death is easily quantified after 7-14 days of reperfusion, as a near complete loss of CA1 neurons is evident at this time. In addition to this brain injury model, we present a method for CA1 damage quantification using a simple, yet thorough, methodology. Importantly, quantification can be accomplished using a simple camera-mounted microscope, and a free ImageJ (NIH) software plugin, obviating the need for cost-prohibitive stereology software programs and a motorized microscopic stage for damage assessment.
Collapse
Affiliation(s)
- Thomas H Sanderson
- Department of Emergency Medicine, Wayne State University School of Medicine.
| | | |
Collapse
|
27
|
Huang SS, Lu YJ, Huang JP, Wu YT, Day YJ, Hung LM. The essential role of endothelial nitric oxide synthase activation in insulin-mediated neuroprotection against ischemic stroke in diabetes. J Vasc Surg 2013; 59:483-91. [PMID: 23663869 DOI: 10.1016/j.jvs.2013.03.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 03/06/2013] [Accepted: 03/15/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Stroke patients with diabetes have a higher mortality rate, worse neurologic outcome, and more severe disability than those without diabetes. Results from clinical trials comparing the outcomes of stroke seen with intensive glycemic control in diabetic individuals are conflicting. Therefore, the present study was aimed to identify the key factor involved in the neuroprotective action of insulin beyond its hypoglycemic effects in streptozotocin-diabetic rats with ischemic stroke. METHODS Long-Evans male rats were divided into three groups (control, diabetes, and diabetes treated with insulin) and subjected to focal cerebral ischemia-reperfusion (FC I/R) injury. RESULTS Hyperglycemia aggravated FC I/R injuries with an increase in cerebral infarction and neurologic deficits, inhibition of glucose uptake and membrane-trafficking activity of glucose transporter 1, and reduction of Akt and endothelial nitric oxide synthase (eNOS) phosphorylation in the cerebrum. Insulin treatment alleviated hyperglycemia and the symptoms of diabetes in streptozotocin-diabetic rats. Insulin administration also significantly decreased cerebral infarction and neurologic deficits and increased phosphorylation of Akt and eNOS protein in the cerebrum of FC I/R-injured diabetic rats. However, the glucose uptake and membrane trafficking activity of glucose transporter 1 in the cerebrum were not restored by insulin treatment. Coadministration of the eNOS inhibitor, N-iminoethyl-L-ornithine, with insulin abrogated beneficial effects of insulin on cerebral infarct volume and neurologic deficits in FC I/R-injured diabetic rats without affecting the hypoglycemic action of insulin. CONCLUSIONS These results suggest that eNOS activation is required for the neuroprotection of insulin against ischemic stroke in patients with diabetes.
Collapse
Affiliation(s)
- Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, Chung Shan Medical University and Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Jhu Lu
- Department and Graduate Institute of Biomedical Sciences and Healthy Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Jiung-Pang Huang
- Department and Graduate Institute of Biomedical Sciences and Healthy Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Yang-Tzu Wu
- Department and Graduate Institute of Biomedical Sciences and Healthy Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Yuan-Ji Day
- Graduate Institute of Clinical Medical Sciences, Chang Gung University and Department of Anesthesiology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Li-Man Hung
- Department and Graduate Institute of Biomedical Sciences and Healthy Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
28
|
Lagina AT, Calo L, Deogracias M, Sanderson T, Kumar R, Wider J, Sullivan JM. Combination therapy with insulin-like growth factor-1 and hypothermia synergistically improves outcome after transient global brain ischemia in the rat. Acad Emerg Med 2013; 20:344-51. [PMID: 23701341 DOI: 10.1111/acem.12104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/25/2012] [Accepted: 10/12/2012] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Hypothermia has a well-established neuroprotective effect and offers a foundation for combination therapy for brain ischemia. The authors evaluated the effect of combination therapy with insulin-like growth factor-1 (IGF-1) and hypothermia on brain structure and function in the setting of global brain ischemia and reperfusion in rats. METHODS Male Sprague-Dawley rats were randomly assigned to groups by a registrar. Animals were subjected to 8 minutes of global brain ischemia using bilateral carotid occlusion and systemic hypotension, followed by 7 days (Stage I dose studies) or 28 days (Stage II outcome studies) of reperfusion. Sham controls were subjected to surgery, but not ischemia. Stage II animals were randomized to no treatment, IGF-1 at the dose determined in Stage I, hypothermia (32°C for 4 hours), or a combination of IGF-1 and hypothermia. Stage II animals underwent 21 days of spatial memory testing. At 7 days (Stage I) or 28 days (Stage II), brains were harvested for counting of CA1 neurons. The primary Stage II outcome was a neurologic outcome index computed as the ratio of viable CA1 neurons per 300-μm field to the number of days to reach success criteria on the memory task. RESULTS Stage I experiments confirmed the neuroprotective effect of the hypothermia protocol and IGF-1 at a dose of 0.6 U/kg. Stage II studies suggested that early neuroprotection with hypothermia and IGF-1 was not well maintained to 28 days and that combination therapy was more beneficial than either IGF-1 or hypothermia alone. Median and interquartile ranges (IQRs) of viable neurons per 300-μm field were 114 (IQR = 99.5 to 136) for sham, three (IQR = 2 to 4.8) for untreated ischemia, four (IQR = 3 to 70.25) for ischemia treated with IGF-1 alone, 25 (IQR = 3 to 70) for ischemia treated with hypothermia alone, and 78 (IQR 47.3 to 97.5) for ischemia treated with combination therapy. Days to memory success criteria were 13.6 (IQR = 11.5 to 15.5 days) for sham, 23.5 (IQR = 20 to 25.5 days) for untreated ischemia, 17.5 (IQR = 15.5 to 25.5 days) for ischemia treated with IGF-1, 15 (IQR = 14.5 to 21 days) for ischemia treated with hypothermia, and 13.5 (IQR = 12.25 to 18.5 days) for ischemia treated with combination therapy. Neurologic outcome indices were 8.5 (IQR = 7.4 to 9.5) for sham, 0.14 (IQR = 0.08 to 0.2) for untreated ischemia, 0.18 (IQR = 0.17 to 4.6) for ischemia treated with IGF-1, 0.7 (IQR = 0.2 to 4.8) for ischemia treated with hypothermia, and 5.7 (IQR = 3.3 to 6.2) for ischemia treated with combination therapy. Statistically significant differences in neuron counts, days to memory test criteria, and outcome index were found between sham and untreated ischemic animals. Of the three treatment regimens, only combination therapy showed a statistically significant difference from the untreated ischemic group for neuronal salvage (p = 0.02), days to criteria (p = 0.043), and outcome index (p = 0.014). CONCLUSIONS Combination therapy with IGF-1 (0.6 U/kg) and therapeutic hypothermia (32°C for 4 hours) at the onset of reperfusion synergistically preserves CA1 structure and function at 28 days after 8 minutes of global brain ischemia in healthy male rats.
Collapse
Affiliation(s)
| | - Lesley Calo
- Cerebral Resuscitation Laboratory; Department of Emergency Medicine; Wayne State University College of Medicine; Detroit; MI
| | - Michael Deogracias
- Cerebral Resuscitation Laboratory; Department of Emergency Medicine; Wayne State University College of Medicine; Detroit; MI
| | | | | | - Joe Wider
- Cerebral Resuscitation Laboratory; Department of Emergency Medicine; Wayne State University College of Medicine; Detroit; MI
| | | |
Collapse
|
29
|
Sanderson TH, Reynolds CA, Kumar R, Przyklenk K, Hüttemann M. Molecular mechanisms of ischemia-reperfusion injury in brain: pivotal role of the mitochondrial membrane potential in reactive oxygen species generation. Mol Neurobiol 2012; 47:9-23. [PMID: 23011809 DOI: 10.1007/s12035-012-8344-z] [Citation(s) in RCA: 465] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/27/2012] [Indexed: 12/20/2022]
Abstract
Stroke and circulatory arrest cause interferences in blood flow to the brain that result in considerable tissue damage. The primary method to reduce or prevent neurologic damage to patients suffering from brain ischemia is prompt restoration of blood flow to the ischemic tissue. However, paradoxically, restoration of blood flow causes additional damage and exacerbates neurocognitive deficits among patients who suffer a brain ischemic event. Mitochondria play a critical role in reperfusion injury by producing excessive reactive oxygen species (ROS) thereby damaging cellular components, and initiating cell death. In this review, we summarize our current understanding of the mechanisms of mitochondrial ROS generation during reperfusion, and specifically, the role the mitochondrial membrane potential plays in the pathology of cerebral ischemia/reperfusion. Additionally, we propose a temporal model of ROS generation in which posttranslational modifications of key oxidative phosphorylation (OxPhos) proteins caused by ischemia induce a hyperactive state upon reintroduction of oxygen. Hyperactive OxPhos generates high mitochondrial membrane potentials, a condition known to generate excessive ROS. Such a state would lead to a "burst" of ROS upon reperfusion, thereby causing structural and functional damage to the mitochondria and inducing cell death signaling that eventually culminate in tissue damage. Finally, we propose that strategies aimed at modulating this maladaptive hyperpolarization of the mitochondrial membrane potential may be a novel therapeutic intervention and present specific studies demonstrating the cytoprotective effect of this treatment modality.
Collapse
Affiliation(s)
- Thomas H Sanderson
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
30
|
Zhao S, Fu J, Liu X, Wang T, Zhang J, Zhao Y. Activation of Akt/GSK-3beta/beta-catenin signaling pathway is involved in survival of neurons after traumatic brain injury in rats. Neurol Res 2012; 34:400-7. [PMID: 22643085 DOI: 10.1179/1743132812y.0000000025] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Apoptotic cell death is an important factor influencing the prognosis after traumatic brain injury (TBI). Akt/GSK-3beta/beta-catenin signaling plays a critical role in the apoptosis of neurons in several models of neurodegeneration. The goal of this study was to determine if the mechanism of cell survival mediated by the Akt/GSK-3beta/beta-catenin pathway is involved in a rat model of TBI. METHODS TBI was performed by a controlled cortical impact device. Expression of Akt, phospho-Akt, GSK-3beta, phospho-GSK-3beta, beta-catenin, phospho-beta-catenin were examined by immunohistochemistry and Western blot analysis. Double immunofluorenscent staining was used to observe the neuronal expression of the aforementioned subtrates. Terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL) staining was performed to identify apoptosis. RESULTS Western blot analysis showed that phospho-Akt significantly increased at 4 hours post-TBI, but decreased after 72 hours post-TBI. Phospho-GSK-3beta - phosphorylated by phospho-Akt - slightly increased at 4 hours post-TBI and peaked at 72 hours post-TBI. These changes in Phospho-GSK-3beta expression were accompanied by a marked increase in expression of phospho-beta-catenin at 4 hours post-TBI which was sustained until 7 days post-TBI. Double staining of phospho-Akt and NeuN revealed the colocalization of phospho-Akt positive cells and neuronal cells. In addition, double staining of phospho-Akt and TUNEL showed no colocalization of phospho-Akt cells and TUNEL-positive cells. CONCLUSION Phosphorylation of Akt (Ser473) and GSK3beta (Ser9) was accelerated in the injured cortex, and involved in the neuronal survival after TBI. Moreover, neuroprotection of beta-catenin against ischemia was partly mediated by enhanced and persistent activation of the Akt/GSK3beta signaling pathway.
Collapse
Affiliation(s)
- Shangfeng Zhao
- Department of Neurosurgery, Beijing Tongren Hospital, Capital University of Medical Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
31
|
The dietary flavonoids naringenin and quercetin acutely impair glucose metabolism in rodents possibly via inhibition of hypothalamic insulin signalling. Br J Nutr 2012; 109:1040-51. [DOI: 10.1017/s0007114512003005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Secondary metabolites of herbs and spices are widely used as an alternative strategy in the therapy of various diseases. The polyphenols naringenin, quercetin and curcumin have been characterised as anti-diabetic agents. Conversely, in vitro, naringenin and quercetin are described to inhibit phosphoinositide-3-kinase (PI3K), an enzyme that is essential for the neuronal control of whole body glucose homoeostasis. Using both in vitro and in vivo experiments, we tested whether the inhibitory effect on PI3K occurs in neurons and if it might affect whole body glucose homoeostasis. Quercetin was found to inhibit basal and insulin-induced phosphorylation of Akt (Ser473), a downstream target of PI3K, in HT-22 cells, whereas naringenin and curcumin had no effect. In Djungarian hamsters (Phodopus sungorus) naringenin and quercetin (10 mg/kg administered orally) diminished insulin-induced phosphorylation of Akt (Ser473) in the arcuate nucleus, indicating a reduction in hypothalamic PI3K activity. In agreement with this finding, glucose tolerance in naringenin-treated hamsters (oral) and mice (oral and intracerebroventricular) was reduced compared with controls. Dietary quercetin also impaired glucose tolerance, whereas curcumin was ineffective. Circulating levels of insulin and insulin-like growth factor-binding protein were not affected by the polyphenols. Oral quercetin reduced the respiratory quotient, suggesting that glucose utilisation was impaired after treatment. These data demonstrate that low doses of naringenin and quercetin acutely and potently impair glucose homoeostasis. This effect may be mediated by inhibition of hypothalamic PI3K signalling. Whether chronic impairments in glucose homoeostasis occur after long-term application remains to be identified.
Collapse
|
32
|
Kim B, McLean LL, Philip SS, Feldman EL. Hyperinsulinemia induces insulin resistance in dorsal root ganglion neurons. Endocrinology 2011; 152:3638-47. [PMID: 21810948 PMCID: PMC3176655 DOI: 10.1210/en.2011-0029] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Insulin resistance (IR) is the major feature of metabolic syndrome, including type 2 diabetes. IR studies are mainly focused on peripheral tissues, such as muscle and liver. There is, however, little knowledge about IR in neurons. In this study, we examined whether neurons develop IR in response to hyperinsulinemia. We first examined insulin signaling using adult dorsal root ganglion neurons as a model system. Acute insulin treatment resulted in time- and concentration-dependent activation of the signaling cascade, including phosphorylation of the insulin receptor, Akt, p70S6K, and glycogen synthase kinase-3β. To mimic hyperinsulinemia, cells were pretreated with 20 nM insulin for 24 h and then stimulated with 20 nM insulin for 15 min. Chronic insulin treatment resulted in increased basal Akt phosphorylation. More importantly, acute insulin stimulation after chronic insulin treatment resulted in blunted phosphorylation of Akt, p70S6K, and glycogen synthase kinase-3β. Interestingly, when the cells were treated with phosphatidylinositol 3-kinase pathway inhibitor, but not MAPK pathway inhibitor, chronic insulin treatment did not block acute insulin treatment-induced Akt phosphorylation. Insulin-induced Akt phosphorylation was lower in dorsal root ganglion neurons from BKS-db/db compared with control BKS-db+ mice. This effect was age dependent. Our results suggest that hyperinsulinemia cause IR by disrupting the Akt-mediated pathway. We also demonstrate that hyperinsulinemia increases the mitochondrial fission protein dynamin-related protein 1. Our results suggest a new theory for the etiology of diabetic neuropathy, i.e. that, similar to insulin dependent tissues, neurons develop IR and, in turn, cannot respond to the neurotrophic properties of insulin, resulting in neuronal injury and the development of neuropathy.
Collapse
Affiliation(s)
- Bhumsoo Kim
- University of Michigan, Department of Neurology, 109 Zina Pitcher Place, 5371 BSRB, Ann Arbor, Michigan 48109-2200, USA.
| | | | | | | |
Collapse
|
33
|
Park JH, Lee CH, Yoo KY, Choi JH, Hwang IK, Lee JY, Kang IJ, Won MH. FoxO3a immunoreactivity is markedly decreased in the dentate gyrus, not the hippocampus proper, of the aged gerbil. Exp Gerontol 2011; 46:836-40. [PMID: 21718780 DOI: 10.1016/j.exger.2011.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 05/23/2011] [Accepted: 06/14/2011] [Indexed: 10/18/2022]
Abstract
Forkhead box O 3a (FoxO3a) has been known to link with aging process and senescence. In this study, we investigated the age-related changes of FoxO3a in the gerbil hippocampus using immunohistochemistry and western blot analysis. In the postnatal month 3 (PM 3) group, FoxO3a immunoreactivity was well detected in pyramidal cells of the hippocampus proper, and granule cells of the dentate gyrus. FoxO3a immunoreactivity in the pyramidal cells of the hippocampus proper was not changed until PM 24. However, in the dentate granule cells, FoxO3a immunoreactivity was much decreased in the dorsal blade, not the ventral blade, of the granule cell layer in the PM 6 and 12 groups compared to the PM 3 group. At PM 24, FoxO3a immunoreactivity in the granule cells was hardly detected. Western blot analysis showed that FoxO3a level was significantly decreased in the PM 24 group. These results indicate that FoxO3a immunoreactivity and levels are markedly decreased in the dentate gyrus of the aged gerbil hippocampus.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim B, Sullivan KA, Backus C, Feldman EL. Cortical neurons develop insulin resistance and blunted Akt signaling: a potential mechanism contributing to enhanced ischemic injury in diabetes. Antioxid Redox Signal 2011; 14:1829-39. [PMID: 21194385 PMCID: PMC3078499 DOI: 10.1089/ars.2010.3816] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Patients with diabetes are at higher risk of stroke and experience increased morbidity and mortality after stroke. We hypothesized that cortical neurons develop insulin resistance, which decreases neuroprotection via circulating insulin and insulin-like growth factor-I (IGF-I). Acute insulin treatment of primary embryonic cortical neurons activated insulin signaling including phosphorylation of the insulin receptor, extracellular signal-regulated kinase (ERK), Akt, p70S6K, and glycogen synthase kinase-3β (GSK-3β). To mimic insulin resistance, cortical neurons were chronically treated with 25 mM glucose, 0.2 mM palmitic acid (PA), or 20 nM insulin before acute exposure to 20 nM insulin. Cortical neurons pretreated with insulin, but not glucose or PA, exhibited blunted phosphorylation of Akt, p70S6K, and GSK-3β with no change detected in ERK. Inhibition of the phosphatidylinositol 3-kinase (PI3-K) pathway during insulin pretreatment restored acute insulin-mediated Akt phosphorylation. Cortical neurons in adult BKS-db/db mice exhibited higher basal Akt phosphorylation than BKS-db(+) mice and did not respond to insulin. Our results indicate that prolonged hyperinsulinemia leads to insulin resistance in cortical neurons. Decreased sensitivity to neuroprotective ligands may explain the increased neuronal damage reported in both experimental models of diabetes and diabetic patients after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | | | | | | |
Collapse
|
35
|
In vivo contributions of BH3-only proteins to neuronal death following seizures, ischemia, and traumatic brain injury. J Cereb Blood Flow Metab 2011; 31:1196-210. [PMID: 21364604 PMCID: PMC3099642 DOI: 10.1038/jcbfm.2011.26] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Bcl-2 homology (BH) domain 3-only proteins are a proapoptotic subgroup of the Bcl-2 gene family, which regulate cell death via effects on mitochondria. The BH3-only proteins react to various cell stressors and promote cell death by binding and inactivating antiapoptotic Bcl-2 family members and direct activation of proapoptotic multi-BH domain proteins such as Bax. Here, we review the in vivo evidence for their involvement in the pathophysiology of status epilepticus and contrast it to ischemia and traumatic brain injury. Seizures in rodents activate three potent proapoptotic BH3-only proteins: Bid, Bim, and Puma. Analysis of damage after seizures in mice singly deficient for each BH3-only protein supports a causal role for Puma and to a lesser extent Bim but, surprisingly, not Bid. In ischemia and trauma, where core aspects of the pathophysiology of cell death overlap, multiple BH3-only proteins are also activated and Bid has been shown to be required for neuronal death. The findings suggest that while each neurologic insult activates multiple BH3-only proteins, there may be specificity in their functional contribution. Future challenges include evaluating the remaining BH3-only proteins, explaining different causal contributions, and, if possible, exploring neurologic outcomes in mouse models deficient for multiple BH3-only proteins.
Collapse
|
36
|
Properties and regulation of organic cation transport in freshly isolated mouse proximal tubules analyzed with a fluorescence reader-based method. Pflugers Arch 2011; 462:359-69. [PMID: 21523352 DOI: 10.1007/s00424-011-0969-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 04/06/2011] [Accepted: 04/11/2011] [Indexed: 01/08/2023]
Abstract
The main elimination site of organic cations (OCs) is the renal proximal tubule (PT). OC transporters (OCT) accept endogenous and exogenous substances and xenobiotics. As transgenic mouse models are increasingly used in translational medicine, functional properties with special focus on regulation of OCT of isolated mouse PTs were studied with a new fluorescence reader-based method, which allows studying larger numbers of tubules per kidney. OC transport across the basolateral membrane of PTs from male mice was measured as initial uptake of the fluorescent dye 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP). A microtiter plate fluorescence reader was used to semi-automatically analyze OC transport in freshly isolated tubules. Relative mRNA expression of OCT1/OCT2/OCT3 in PTs was 1/0.3/0.01 and did not vary from S1 to S3 segments. ASP was transported by PTs with a K (m) of 6 μM. It was inhibited by TEA, TPA, or cimetidine (IC(50)=5, 19, or 53 μM, respectively). Angiotensin II stimulated ASP uptake (+63%), while stimulation of PKC reduced (-37%) OC transport. Inhibition of p56(lck) tyrosine kinase (-60%), of PI3K (-36%), of Ca(2+)/calmodulin (-25%), or of PKA (-33%) reduced OC transport. In PTs from OCT1/2(-/-) mice ASP uptake was reduced to ~20%. Using this fluorescence reader-based method, we report substrate specificities and a complex pattern of acute regulation of OC transport in isolated mouse PTs. Compared to isolated human PTs or rat and human OCT isoforms expressed in HEK293-cells, OC transport across the basolateral membrane of freshly isolated mouse PTs shows similarities but also specific differences.
Collapse
|
37
|
Mielke JG, Wang YT. Insulin, synaptic function, and opportunities for neuroprotection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:133-86. [PMID: 21199772 DOI: 10.1016/b978-0-12-385506-0.00004-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A steadily growing number of studies have begun to establish that the brain and insulin, while traditionally viewed as separate, do indeed have a relationship. The uptake of pancreatic insulin, along with neuronal biosynthesis, provides neural tissue with the hormone. As well, insulin acts upon a neuronal receptor that, although a close reflection of its peripheral counterpart, is characterized by unique structural and functional properties. One distinction is that the neural variant plays only a limited part in neuronal glucose transport. However, a number of other roles for neural insulin are gradually emerging; most significant among these is the modulation of ligand-gated ion channel (LGIC) trafficking. Notably, insulin has been shown to affect the tone of synaptic transmission by regulating cell-surface expression of inhibitory and excitatory receptors. The manner in which insulin regulates receptor movement may provide a cellular mechanism for insulin-mediated neuroprotection in the absence of hypoglycemia and stimulate the exploration of new therapeutic opportunities.
Collapse
Affiliation(s)
- John G Mielke
- Faculty of Applied Health Sciences, Department of Health Studies and Gerontology, University of Waterloo, Waterloo, Ontario, Canada
| | | |
Collapse
|
38
|
den Hengst WA, Gielis JF, Lin JY, Van Schil PE, De Windt LJ, Moens AL. Lung ischemia-reperfusion injury: a molecular and clinical view on a complex pathophysiological process. Am J Physiol Heart Circ Physiol 2010; 299:H1283-99. [PMID: 20833966 DOI: 10.1152/ajpheart.00251.2010] [Citation(s) in RCA: 274] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lung ischemia-reperfusion injury remains one of the major complications after cardiac bypass surgery and lung transplantation. Due to its dual blood supply system and the availability of oxygen from alveolar ventilation, the pathogenetic mechanisms of ischemia-reperfusion injury in the lungs are more complicated than in other organs, where loss of blood flow automatically leads to hypoxia. In this review, an extensive overview is given of the molecular and cellular mechanisms that are involved in the pathogenesis of lung ischemia-reperfusion injury and the possible therapeutic strategies to reduce or prevent it. In addition, the roles of neutrophils, alveolar macrophages, cytokines, and chemokines, as well as the alterations in the cell-death related pathways, are described in detail.
Collapse
Affiliation(s)
- Willem A den Hengst
- Department of Thorax and Vascular Surgery, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
39
|
Al-Shabrawey M, Smith S. Prediction of diabetic retinopathy: role of oxidative stress and relevance of apoptotic biomarkers. EPMA J 2010; 1:56-72. [PMID: 23199041 PMCID: PMC3405307 DOI: 10.1007/s13167-010-0002-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 01/25/2010] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy (DR) is the foremost cause of blindness in working-aged worldwide; it is characterized by vascular and neuronal degeneration. Features of DR include leukocyte adhesion, increased vascular permeability, neovascularization and neuronal cell death. Early diagnosis and intervention are important to prevent or at least ameliorate the development of DR. Recent reports indicate that pathophysiological mechanisms leading to diabetic retinopathy include oxidative stress and retinal cell death cascades. Circulating biomarkers of oxidative stress such as malondialdehyde (MDA), thiobarbituric acid reacting substances (TBARS), conjugated diene (CD), advanced oxidation protein products (AOPP), protein carbonyl, 8-hydroxydeoxyguanosin (8-OHdG), nitrotyrosine, and F(2) isoprostanes and pro-apoptosis molecules (caspase-3, Fas, and Bax) are associated with increased susceptibility to develop DR in diabetic subjects. Thus, identification of oxidative stress and cell death biomarkers in diabetic patients could be in favor of predicting, diagnosis, and prevention of DR, and to target for novel therapeutic interventions.
Collapse
Affiliation(s)
- Mohamed Al-Shabrawey
- Oral Biology and Anatomy, School of Dentistry, Medical College of Georgia, Augusta, GA 30912 USA
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, Augusta, GA 30912 USA
- Opthalmology, King Saud University, Riyadh, Saudi Arabia
| | - Sylvia Smith
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, Augusta, GA 30912 USA
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912 USA
| |
Collapse
|
40
|
Cui M, Huang Y, Zhao Y, Zheng J. New insights for FOXO and cell-fate decision in HIV infection and HIV associated neurocognitive disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 665:143-59. [PMID: 20429422 DOI: 10.1007/978-1-4419-1599-3_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human immunodeficiency virus Type 1 (HIV-1) infection and associated diseases continue to represent major health problem worldwide. FOXO transcriptional factors play an important role in the regulation of cell apoptosis, cell cycle arrest, stress resistance, metabolism and differentiation. This chapter will discuss the diverse functions of FOXO in different cell types including T-cells, macrophages, neurons and astrocytes within the context of HIV-1 infection. Given the overwhelming evidence that FOXO proteins influence the cell fate of immune cells and involve in the homeostasis of the central nervous system (CNS), we will also discuss the potential role of FOXO factors in HIV-1-associated neurological disorders.
Collapse
Affiliation(s)
- Min Cui
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|