1
|
Arslan R, Doganay S, Budak O, Bahtiyar N. Investigation of preconditioning and the protective effects of nicotinamide against cerebral ischemia-reperfusion injury in rats. Neurosci Lett 2024; 840:137949. [PMID: 39181500 DOI: 10.1016/j.neulet.2024.137949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/04/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This study investigated the antioxidant and neuroprotective effects of nicotinamide combined with ischemic preconditioning against cerebral ischemia reperfusion (CIR) injury. Thirty-five Wistar albino male rats were randomly divided into five groups: sham, preconditioned ischemia/reperfusion (IP+IR), ischemia/reperfusion (IR), preconditioned ischemia/reperfusion + nicotinamide (IP+IR+N), and ischemia/reperfusion + nicotinamide (IR+N). CIR was achieved with bilateral common carotid artery occlusion. IP+IR and IP+IR+N groups 30 min before ischemia; Three cycles of 10 sec ischemia/30 sec reperfusion followed by 20 min IR were applied. The IP+IR+N and IR+N groups received 500 mg/kg nicotinamide intraperitoneally. After 24 h of reperfusion, a neurological evaluation was performed and vertıcal pole test. Biochemically, malondialdehyde (MDA), glutathione (GSH) levels and catalase (CAT) activity were measured in blood and brain tissue samples. Rates of red neurons, sateliosis and spongiosis were determined histopathologically in the prefrontal cortex areas. After CIR, MDA levels increased significantly in serum and brain tissue in the IR group compared to the sham group, while GSH and CAT activity decreased in the brain tissue (p < 0.05). MDA levels in the tissues were found significantly decreased in the IR+N group compared to the IR group (p < 0.05). Administration of nicotinamide together with IP significantly decreased MDA levels in brain tissue and increased GSH and CAT activity (p < 0.05). Compared to the IR group, the morphological and neurological damage in the prefrontal cortex areas decreased in the IP+IR, IP+IR+N, and IR+N groups (p < 0.05). In addition, red neuron, sateliosis and spongiosis rates increased significantly in the IR group compared to the Sham, IP+IR+N, IR+N groups (p < 0.001 for all). In neurological evaluation, while the neurological score increased and the time on the vertical pole decreased significantly in the IR group, preconditioning, and nicotinamide groups reversed (p < 0.05). The study's results show that nicotinamide administration with ischemic preconditioning alleviates cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ruhat Arslan
- Istinye University, Faculty of Medicine, Department of Physiology, TR - 34000 Istanbul, Turkey; Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, TR-34098 Istanbul, Turkey.
| | - Songul Doganay
- Sakarya University, Faculty of Medicine, Department of Physiology, TR-54000 Sakarya, Turkey.
| | - Ozcan Budak
- Sakarya University, Faculty of Medicine, Department of Histology and Embryology, TR-54000 Sakarya, Turkey.
| | - Nurten Bahtiyar
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Biophysics, TR-34098 Istanbul, Turkey.
| |
Collapse
|
2
|
ZHANG X, GUO D, ZHANG X, ZHANG W, WANG T, ZHANG L. Three-N-butyphthalide alleviates early brain injury caused via subarachnoid hemorrhage via activating the LKB-1/ (AMP-activated protein kinase) pathway. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.86321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
3
|
AMPK inhibitor BML-275 induces neuroprotection through decreasing cyt c and AIF expression after transient brain ischemia. Bioorg Med Chem 2021; 52:116522. [PMID: 34837819 DOI: 10.1016/j.bmc.2021.116522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022]
Abstract
Stroke is a major public health problem with an imperative need for a more effective and tolerated therapy. Neuroprotective therapy may be an effective therapeutic intervention for stroke. The morbidity and mortality of stroke-induced secondary brain injury is mainly caused by neuronal apoptosis, which can be executed in a caspase-dependent or apoptosis inducing factor (AIF)-dependent manner. As apoptosis is an energy-dependent process with a relative time delay, abnormal energy metabolism could be a significant and fundamental pathophysiological basis of stroke. To our knowledge, convincible evidences that AMPK inhibition exerts neuroprotection in cerebral ischemia injury via anti-apoptosis remain to be investigated. Accordingly, the aims of this study were to investigate the protective effects of AMPK inhibitor BML-275 on cerebral ischemic/reperfusion (I/R) injury and to elucidate the underlying mechanisms. Cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) in male C57BL/6 mice. The therapeutic effects of BML-275 were evaluated by infarct sizes, neurological scores and the proportion of apoptotic neurons after 24 h of reperfusion. The cell apoptosis markers cyt c and AIF were also evaluated. The results showed that intraperitoneally administration of BML-275 alleviate the cerebral infarction, neurological deficit and neuronal apoptosis induced by MCAO. BML-275 simultaneously induces anti-apoptosis and decreases the expression of cyt c and AIF. This study supports the hypothesis that anti-apoptosis is one of potential neuroprotective strategies for the treatment of stroke.
Collapse
|
4
|
Pfeiffer S, Tomašcová A, Mamrak U, Haunsberger SJ, Connolly NMC, Resler A, Düssmann H, Weisová P, Jirström E, D'Orsi B, Chen G, Cremona M, Hennessy BT, Plesnila N, Prehn JHM. AMPK-regulated miRNA-210-3p is activated during ischaemic neuronal injury and modulates PI3K-p70S6K signalling. J Neurochem 2021; 159:710-728. [PMID: 33694332 DOI: 10.1111/jnc.15347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/12/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022]
Abstract
Progressive neuronal injury following ischaemic stroke is associated with glutamate-induced depolarization, energetic stress and activation of AMP-activated protein kinase (AMPK). We here identify a molecular signature associated with neuronal AMPK activation, as a critical regulator of cellular response to energetic stress following ischaemia. We report a robust induction of microRNA miR-210-3p both in vitro in primary cortical neurons in response to acute AMPK activation and following ischaemic stroke in vivo. Bioinformatics and reverse phase protein array analysis of neuronal protein expression changes in vivo following administration of a miR-210-3p mimic revealed altered expression of phosphatase and tensin homolog (PTEN), 3-phosphoinositide-dependent protein kinase 1 (PDK1), ribosomal protein S6 kinase (p70S6K) and ribosomal protein S6 (RPS6) signalling in response to increasing miR-210-3p. In vivo, we observed a corresponding reduction in p70S6K activity following ischaemic stroke. Utilizing models of glutamate receptor over-activation in primary neurons, we demonstrated that induction of miR-210-3p was accompanied by sustained suppression of p70S6K activity and that this effect was reversed by miR-210-3p inhibition. Collectively, these results provide new molecular insight into the regulation of cell signalling during ischaemic injury, and suggest a novel mechanism whereby AMPK regulates miR-210-3p to control p70S6K activity in ischaemic stroke and excitotoxic injury.
Collapse
Affiliation(s)
- Shona Pfeiffer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Anna Tomašcová
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Biomedical Centre Martin, Comenius University in Bratislava, Bratislava, Slovakia
| | - Uta Mamrak
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Stefan J Haunsberger
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Niamh M C Connolly
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alexa Resler
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Düssmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Petronela Weisová
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elisabeth Jirström
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Center, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Beatrice D'Orsi
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Institute of Neuroscience, Italian National Research Council (CNR), Pisa, Italy
| | - Gang Chen
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mattia Cremona
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Dept of Molecular Medicine (Medical Oncology group), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Dept of Molecular Medicine (Medical Oncology group), Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Center, Royal College of Surgeons Ireland, Dublin, Ireland
| |
Collapse
|
5
|
Stacey BS, Campbell Z, Bailey DM. Elevated cerebral perfusion and preserved cognition in elite Brazilian Jiu-Jitsu athletes: Evidence for neuroprotection. Scand J Med Sci Sports 2021; 31:2115-2122. [PMID: 34343371 DOI: 10.1111/sms.14031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 12/01/2022]
Abstract
Brazilian Jiu-Jitsu (BJJ) is a popular martial art that exposes participants to recurrent intermittent asphyxiation due to controlled application of neck chokes. To what extent the sport impacts the regulation of cerebral blood flow (CBF) and cognition has not been examined. This study compared eleven elite Brazilian Jiu-Jitsu athletes (aged 30 ± 8 y) who trained 12 ± 6 hours/week for 8 ± 4 years against eleven cardiorespiratory fitness (CRF)- and age-matched controls. Internal carotid (ICA) and vertebral artery (VA) blood flow were measured via duplex ultrasound to determine global cerebral blood flow (gCBF). Mild cognitive impairment and sub-domains of memory, attention/concentration/visual motor coordination, and executive function were determined by psychometric testing. There was no evidence of mild cognitive impairment in the athletes, and cognitive function was comparable between groups (all p > 0.05). In contrast, resting gCBF was selectively elevated in the athletes (741 ± 186 mL∙min-1 vs. 573 ± 166 mL∙min-1 , p = 0.037) due to combined differences in ICA (+65 mL∙min-1 , p = 0.079) and VA (+19 mL∙min-1 , p = 0.277) flow. In conclusion, the sustained elevation in resting cerebral perfusion provides preliminary evidence for adaptive neuroprotection that is independent of CRF and likely mediated by choke-induced cerebral preconditioning and/or lifelong exposure to BJJ-specific high-intensity interval training.
Collapse
Affiliation(s)
- Benjamin S Stacey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, UK
| | - Zac Campbell
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, UK
| | - Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, UK
| |
Collapse
|
6
|
Deficiency of ROS-Activated TRPM2 Channel Protects Neurons from Cerebral Ischemia-Reperfusion Injury through Upregulating Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7356266. [PMID: 34367466 PMCID: PMC8337124 DOI: 10.1155/2021/7356266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022]
Abstract
Cerebral ischemia-reperfusion (I-R) transiently increased autophagy by producing excessively reactive oxygen species (ROS); on the other hand, activated autophagy would remove ROS-damaged mitochondria and proteins, which led to cell survival. However, the regulation mechanism of autophagy activity during cerebral I-R is still unclear. In this study, we found that deficiency of the TRPM2 channel which is a ROS sensor significantly decreased I-R-induced neuronal damage. I-R transiently increased autophagy activity both in vitro and in vivo. More importantly, TRPM2 deficiency decreased I-R-induced neurological deficit score and infarct volume. Interestingly, our results indicated that TRPM2 deficiency could further activate AMPK rather than Beclin1 activity, suggesting that TRPM2 inhibits autophagy by regulating the AMPK/mTOR pathway in I-R. In conclusion, our study reveals that ROS-activated TRPM2 inhibits autophagy by downregulating the AMPK/mTOR pathway, which results in neuronal death induced by cerebral I-R, further supporting that TRPM2 might be a potential drug target for cerebral ischemic injury therapy.
Collapse
|
7
|
Hao Y, Xin M, Feng L, Wang X, Wang X, Ma D, Feng J. Review Cerebral Ischemic Tolerance and Preconditioning: Methods, Mechanisms, Clinical Applications, and Challenges. Front Neurol 2020; 11:812. [PMID: 33071923 PMCID: PMC7530891 DOI: 10.3389/fneur.2020.00812] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide, and it is increasing in prevalence. The limited therapeutic window and potential severe side effects prevent the widespread clinical application of the venous injection of thrombolytic tissue plasminogen activator and thrombectomy, which are regarded as the only approved treatments for acute ischemic stroke. Triggered by various types of mild stressors or stimuli, ischemic preconditioning (IPreC) induces adaptive endogenous tolerance to ischemia/reperfusion (I/R) injury by activating a multitude cascade of biomolecules, for example, proteins, enzymes, receptors, transcription factors, and others, which eventually lead to transcriptional regulation and epigenetic and genomic reprogramming. During the past 30 years, IPreC has been widely studied to confirm its neuroprotection against subsequent I/R injury, mainly including local ischemic preconditioning (LIPreC), remote ischemic preconditioning (RIPreC), and cross preconditioning. Although LIPreC has a strong neuroprotective effect, the clinical application of IPreC for subsequent cerebral ischemia is difficult. There are two main reasons for the above result: Cerebral ischemia is unpredictable, and LIPreC is also capable of inducing unexpected injury with only minor differences to durations or intensity. RIPreC and pharmacological preconditioning, an easy-to-use and non-invasive therapy, can be performed in a variety of clinical settings and appear to be more suitable for the clinical management of ischemic stroke. Hoping to advance our understanding of IPreC, this review mainly focuses on recent advances in IPreC in stroke management, its challenges, and the potential study directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Ma HX, Hou F, Chen AL, Li TT, Zhu YF, Zhao QP. Mu-Xiang-You-Fang protects PC12 cells against OGD/R-induced autophagy via the AMPK/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112583. [PMID: 31978519 DOI: 10.1016/j.jep.2020.112583] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/30/2019] [Accepted: 01/15/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mu-Xiang-You-Fang (MXYF) is a classic prescription of Hui medicine. It is composed of five herbs and has been used to treat ischemic stroke for many years. However, the potential pharmacological mechanisms of MXYF remain unclear. The present research is aimed to investigate the protective effect and possible mechanisms of MXYF treatment in an in vitro model of cerebral ischemia-reperfusion injury. MATERIALS AND METHODS An oxygen-glucose deprivation and reperfusion (OGD/R) model of PC12 cells was established. The effect of MXYF on the cell viability after OGD/R injury was determined using a cell counting kit (CCK-8) assay. The colorimetric method was used to determine the lactate dehydrogenase (LDH) leakage rate. The calcium concentration was determined by the chemical fluorescence method, and mitochondrial membrane potential was determined using flow cytometry. Monodansylcadaverine (MDC) staining and electron microscopic analysis were then conducted to detect autophagy after oxygen-glucose deprivation and reperfusion in PC12 cells. Immunofluorescence and western blot analyses were used to detect the expression of proteins associated with autophagy. RESULTS It was found that MXYF (1, 2, 4 μg/mL) could significantly increase cell viability and mitochondrial membrane potential and decrease the calcium concentration and LDH release rate in PC12 cells. After OGD/R injury in PC12 cells, the number of autophagosomes and autophagolysosome significantly increased. MXYF (4 μg/mL) inhibited the autophagy induced by OGD/R and inhibited the expression of LC3, beclin1, p-AMPK, and ULK1. In contrast, the expression of p-mTOR, p-p70s6k, and p62 was significantly enhanced. CONCLUSIONS These findings suggest that MXYF inhibits autophagy after OGD/R-induced PC12 cell injury through the AMPK-mTOR pathway. Thus, MXYF might have therapeutic potential in treating ischemic stroke.
Collapse
Affiliation(s)
- Hui-Xia Ma
- Key Laboratory of Modern Hui Medicine, Ningxia Medical University, Ningxia, China; Department of Pharmacology, Ningxia Medical University, Ningxia, China
| | - Fan Hou
- Key Laboratory of Modern Hui Medicine, Ningxia Medical University, Ningxia, China; Department of Pharmacology, Ningxia Medical University, Ningxia, China
| | - Ai-Ling Chen
- Key Laboratory of Modern Hui Medicine, Ningxia Medical University, Ningxia, China; Department of Pharmacology, Ningxia Medical University, Ningxia, China
| | - Ting-Ting Li
- Key Laboratory of Modern Hui Medicine, Ningxia Medical University, Ningxia, China
| | - Ya-Fei Zhu
- College of Basic Medicine, Ningxia Medical University, Ningxia, China.
| | - Qi-Peng Zhao
- Key Laboratory of Modern Hui Medicine, Ningxia Medical University, Ningxia, China; Department of Pharmacology, Ningxia Medical University, Ningxia, China.
| |
Collapse
|
9
|
Khedr LH, Nassar NN, Rashed L, El-Denshary ED, Abdel-Tawab AM. TLR4 signaling modulation of PGC1-α mediated mitochondrial biogenesis in the LPS-Chronic mild stress model: Effect of fluoxetine and pentoxiyfylline. Life Sci 2019; 239:116869. [PMID: 31678277 DOI: 10.1016/j.lfs.2019.116869] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 11/15/2022]
Abstract
AIM The addition of repeated lipopolysaccharide (LPS) to chronic mild stress was recently proposed in our lab as an alternative model of depression, highlighting the possible interaction between stress and immune-inflammatory pathways in predisposing depression. Given that CMS-induced depressive behavior was previously related to impaired hippocampal energy metabolism and mitochondrial dysfunction, our current study aimed to investigate the interplay between toll-like receptor 4 (TLR4) signaling and peroxisome proliferator-activated receptor gamma coactivators-1-alpha (PGC1-α) as a physiological regulator of energy metabolism and mitochondrial biogenesis in the combined LPS/CMS model. MAIN METHODS Male Wistar rats were exposed to either LPS (50 μg/kg i.p.) over 2 weeks, CMS protocol for 4 weeks or LPS over 2 weeks followed by 4 weeks of CMS (LPS/CMS). Three additional groups of rats were exposed to LPS/CMS protocol and treated with either pentoxifylline (PTX), fluoxetine (FLX) or a combination of both. Rats were examined for behavioral, neurochemical, gene expression and mitochondrial ultra-structural changes. KEY FINDINGS LPS/CMS increased the expression of TLR4 and its downstream players; MyD88, NFκB and TNF-α along with an escalation in hippocampal-energy metabolism and p-AMPK. Simultaneously LPS/CMS attenuated the expression of PGC1-α/NRF1/Tfam and mt-DNA. The antidepressant (AD) 'FLX', the TNF-α inhibitor 'PTX' and their combination ameliorated the LPS/CMS-induced changes. Interestingly, all the aforementioned changes induced by the LPS/CMS combined model were significantly less than those induced by CMS alone. SIGNIFICANCE Blocking the TLR4/NFκB signaling enhanced the activation of the PGC1-α/NRF1/Tfam and mt-DNA content independent on the activation of the energy-sensing kinase AMPK.
Collapse
Affiliation(s)
- L H Khedr
- Departmment of Pharmacology, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| | - N N Nassar
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - E D El-Denshary
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - A M Abdel-Tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
10
|
McDonough A, Noor S, Lee RV, Dodge R, Strosnider JS, Shen J, Davidson S, Möller T, Garden GA, Weinstein JR. Ischemic preconditioning induces cortical microglial proliferation and a transcriptomic program of robust cell cycle activation. Glia 2019; 68:76-94. [PMID: 31420975 DOI: 10.1002/glia.23701] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022]
Abstract
Ischemic preconditioning (IPC) is an experimental phenomenon in which a subthreshold ischemic insult applied to the brain reduces damage caused by a subsequent more severe ischemic episode. Identifying key molecular and cellular mediators of IPC will provide critical information needed to develop novel therapies for stroke. Here we report that the transcriptomic response of acutely isolated preconditioned cortical microglia is dominated by marked upregulation of genes involved in cell cycle activation and cellular proliferation. Notably, this transcriptional response occurs in the absence of cortical infarction. We employed ex vivo flow cytometry, immunofluorescent microscopy, and quantitative stereology methods on brain tissue to evaluate microglia proliferation following IPC. Using cellular colocalization of microglial (Iba1) and proliferation (Ki67 and BrdU) markers, we observed a localized increase in the number of microglia and proliferating microglia within the preconditioned hemicortex at 72, but not 24, hours post-IPC. Our quantification demonstrated that the IPC-induced increase in total microglia was due entirely to proliferation. Furthermore, microglia in the preconditioned hemisphere had altered morphology and increased soma volumes, indicative of an activated phenotype. Using transgenic mouse models with either fractalkine receptor (CX3CR1)-haploinsufficiency or systemic type I interferon signaling loss, we determined that microglial proliferation after IPC is dependent on fractalkine signaling but independent of type I interferon signaling. These findings suggest there are multiple distinct targetable signaling pathways in microglia, including CX3CR1-dependent proliferation that may be involved in IPC-mediated protection.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Shahani Noor
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Richard V Lee
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Ryan Dodge
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - James S Strosnider
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Jasmine Shen
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Stephanie Davidson
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Thomas Möller
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Gwenn A Garden
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington.,Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
11
|
McDonough A, Weinstein JR. The role of microglia in ischemic preconditioning. Glia 2019; 68:455-471. [PMID: 31386233 DOI: 10.1002/glia.23695] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/20/2019] [Accepted: 07/23/2019] [Indexed: 12/22/2022]
Abstract
Ischemic preconditioning (IPC) is an experimental phenomenon in which a brief ischemic stimulus confers protection against a subsequent prolonged ischemic event. Initially thought to be due to mechanistic changes in neurons, our understanding of IPC has evolved to encompass a global reprogramming of the Central Nervous System (CNS) after transient ischemia/reperfusion that requires innate immune signaling pathways including Toll-like receptors (TLRs) and Type I interferons. Microglia are the CNS resident neuroimmune cells that express these key innate immune receptors. Studies suggest that microglia are required for IPC-mediated neuronal and axonal protection. Multiple paradigms targeting TLRs have converged on a distinctive Type I interferon response in microglia that is critical for preconditioning-mediated protection against ischemia. These pathways can be targeted through administration of TLR agonists, cytokines including interferon-β, and pharmaceutical agents that induce preconditioning through cross-tolerance mechanisms. Transcriptomic analyses and single cell RNA studies point to specific gene expression signatures in microglia that functionally shift these mutable cells to an immunomodulatory or protective phenotype. Although there are technological challenges and gaps in knowledge to overcome, the targeting of specific molecular signaling pathways in microglia is a promising direction for development of novel and effective pharmacotherapies for stroke. Studies on preconditioning in animal models, including nonhuman primates, show promise as prophylactic preconditioning treatments for selected at risk patient populations. In addition, our growing understanding of the mechanisms of IPC-mediated protection is identifying novel cellular and molecular targets for therapeutic interventions that could apply broadly to both acute stroke and chronic vascular cognitive impairment patients.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington.,Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
12
|
A L, Zou T, He J, Chen X, Sun D, Fan X, Xu H. Rescue of Retinal Degeneration in rd1 Mice by Intravitreally Injected Metformin. Front Mol Neurosci 2019; 12:102. [PMID: 31080404 PMCID: PMC6497809 DOI: 10.3389/fnmol.2019.00102] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/04/2019] [Indexed: 12/18/2022] Open
Abstract
Retinitis pigmentosa (RP) is a progressive hereditary retinal degenerative disease in which photoreceptor cells undergo degeneration and apoptosis, eventually resulting in irreversible loss of visual function. Currently, no effective treatment exists for this disease. Neuroprotection and inflammation suppression have been reported to delay the development of RP. Metformin is a well-tested drug used to treat type 2 diabetes, and it has been reported to exert beneficial effects in neurodegenerative diseases, such as Parkinson’s disease and Alzheimer’s disease. In the present study, we used immunofluorescence staining, electroretinogram (ERG) recordings and RNA-Seq to explore the effects of metformin on photoreceptor degeneration and its mechanism in rd1 mice. We found that metformin significantly reduced apoptosis in photoreceptors and delayed the degeneration of photoreceptors and rod bipolar cells in rd1 mice, thus markedly improving the visual function of rd1 mice at P14, P18, and P22 when tested with a light/dark transition test and ERG. Microglial activation in the outer nuclear layer (ONL) of the retina of rd1 mice was significantly suppressed by metformin. RNA-Seq showed that metformin markedly downregulated inflammatory genes and upregulated the expression of crystallin proteins, which have been demonstrated to be important neuroprotective molecules in the retina, revealing the therapeutic potential of metformin for RP treatment. αA-crystallin proteins were further confirmed to be involved in the neuroprotective effects of metformin in a Ca2+ ionophore-damaged 661W photoreceptor-like cell line. These data suggest that metformin exerts a protective effect in rd1 mice via both immunoregulatory and new neuroprotective mechanisms.
Collapse
Affiliation(s)
- Luodan A
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China.,Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Ting Zou
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Juncai He
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Xia Chen
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Dayu Sun
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haiwei Xu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
13
|
McDonough A, Weinstein JR. Correction to: Neuroimmune Response in Ischemic Preconditioning. Neurotherapeutics 2018; 15:511-524. [PMID: 29110213 PMCID: PMC5935631 DOI: 10.1007/s13311-017-0580-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Ischemic preconditioning (IPC) is a robust neuroprotective phenomenon in which a brief period of cerebral ischemia confers transient tolerance to subsequent ischemic challenge. Research on IPC has implicated cellular, molecular, and systemic elements of the immune response in this phenomenon. Potent molecular mediators of IPC include innate immune signaling pathways such as Toll-like receptors and type 1 interferons. Brain ischemia results in release of pro- and anti-inflammatory cytokines and chemokines that orchestrate the neuroinflammatory response, resolution of inflammation, and transition to neurological recovery and regeneration. Cellular mediators of IPC include microglia, the resident central nervous system immune cells, astrocytes, and neurons. All of these cell types engage in cross-talk with each other using a multitude of signaling pathways that modulate activation/suppression of each of the other cell types in response to ischemia. As the postischemic neuroimmune response evolves over time there is a shift in function toward provision of trophic support and neuroprotection. Peripheral immune cells infiltrate the central nervous system en masse after stroke and are largely detrimental, with a few subtypes having beneficial, protective effects, though the role of these immune cells in IPC is largely unknown. The role of neural progenitor cells in IPC-mediated neuroprotection is another active area of investigation as is the role of microglial proliferation in this setting. A mechanistic understanding of these molecular and cellular mediators of IPC may not only facilitate more effective direct application of IPC to specific clinical scenarios, but also, more broadly, reveal novel targets for therapeutic intervention in stroke.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
14
|
How AMPK and PKA Interplay to Regulate Mitochondrial Function and Survival in Models of Ischemia and Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4353510. [PMID: 29391924 PMCID: PMC5748092 DOI: 10.1155/2017/4353510] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a conserved, redox-activated master regulator of cell metabolism. In the presence of oxidative stress, AMPK promotes cytoprotection by enhancing the conservation of energy by suppressing protein translation and by stimulating autophagy. AMPK interplays with protein kinase A (PKA) to regulate oxidative stress, mitochondrial function, and cell survival. AMPK and dual-specificity A-kinase anchoring protein 1 (D-AKAP1), a mitochondrial-directed scaffold of PKA, interact to regulate mitochondrial function and oxidative stress in cardiac and endothelial cells. Ischemia and diabetes, a chronic disease that increases the onset of cardiovascular diseases, suppress the cardioprotective effects of AMPK and PKA. Here, we review the molecular mechanisms by which AMPK and D-AKAP1/PKA interplay to regulate mitochondrial function, oxidative stress, and signaling pathways that prime endothelial cells, cardiac cells, and neurons for cytoprotection against oxidative stress. We discuss recent literature showing how temporal dynamics and localization of activated AMPK and PKA holoenzymes play a crucial role in governing cellular bioenergetics and cell survival in models of ischemia, cardiovascular diseases, and diabetes. Finally, we propose therapeutic strategies that tout localized PKA and AMPK signaling to reverse mitochondrial dysfunction, oxidative stress, and death of neurons and cardiac and endothelial cells during ischemia and diabetes.
Collapse
|
15
|
Postconditioning-induced neuroprotection, mechanisms and applications in cerebral ischemia. Neurochem Int 2017; 107:43-56. [DOI: 10.1016/j.neuint.2017.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/04/2017] [Accepted: 01/08/2017] [Indexed: 02/07/2023]
|
16
|
Tian Y, Qi M, Wang Z, Wu C, Sun Z, Li Y, Sha S, Du Y, Chen L, Chen L. Activation of Transient Receptor Potential Vanilloid 4 Impairs the Dendritic Arborization of Newborn Neurons in the Hippocampal Dentate Gyrus through the AMPK and Akt Signaling Pathways. Front Mol Neurosci 2017; 10:190. [PMID: 28663724 PMCID: PMC5471311 DOI: 10.3389/fnmol.2017.00190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/29/2017] [Indexed: 11/13/2022] Open
Abstract
Neurite growth is an important process for the adult hippocampal neurogenesis which is regulated by a specific range of the intracellular free Ca2+ concentration ([Ca2+]i). Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable channel and activation of it causes an increase in [Ca2+]i. We recently reported that TRPV4 activation promotes the proliferation of stem cells in the adult hippocampal dentate gyrus (DG). The present study aimed to examine the effect of TRPV4 activation on the dendrite morphology of newborn neurons in the adult hippocampal DG. Here, we report that intracerebroventricular injection of the TRPV4 agonist GSK1016790A for 5 days (GSK1016790A-injected mice) reduced the number of doublecortin immunopositive (DCX+) cells and DCX+ fibers in the hippocampal DG, showing the impaired dendritic arborization of newborn neurons. The phosphorylated AMP-activated protein kinase (p-AMPK) protein level increased from 30 min to 2 h, and then decreased from 1 to 5 days after GSK1016790A injection. The phosphorylated protein kinase B (p-Akt) protein level decreased from 30 min to 5 days after GSK1016790A injection; this decrease was markedly attenuated by the AMPK antagonist compound C (CC), but not by the AMPK agonist AICAR. Moreover, the phosphorylated mammalian target of rapamycin (mTOR) and p70 ribosomal S6 kinase (p70S6k) protein levels were decreased by GSK1016790A; these changes were sensitive to 740 Y-P and CC. The phosphorylation of glycogen synthase kinase 3β (GSK3β) at Y216 was increased by GSK1016790A, and this change was accompanied by increased phosphorylation of microtubule-associated protein 2 (MAP2) and collapsin response mediator protein-2 (CRMP-2). These changes were markedly blocked by 740 Y-P and CC. Finally, GSK1016790A-induced decrease of DCX+ cells and DCX+ fibers was markedly attenuated by 740 Y-P and CC, but was unaffected by AICAR. We conclude that TRPV4 activation impairs the dendritic arborization of newborn neurons through increasing AMPK and inhibiting Akt to inhibit the mTOR-p70S6k pathway, activate GSK3β and thereby result in the inhibition of MAP2 and CRMP-2 function.
Collapse
Affiliation(s)
- Yujing Tian
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Mengwen Qi
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Zhouqing Wang
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Chunfeng Wu
- Department of Neurology, Children's Hospital of Nanjing Medical UniversityNanjing, China
| | - Zhen Sun
- Department of Tangshan Branch, Jinling Hospital, Nanjing UniversityNanjing, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Sha Sha
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Lei Chen
- Department of Physiology, Nanjing Medical UniversityNanjing, China.,Neuroprotective Drug Discovery Center, Nanjing Medical UniversityNanjing, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| |
Collapse
|
17
|
Arbeláez-Quintero I, Palacios M. To Use or Not to Use Metformin in Cerebral Ischemia: A Review of the Application of Metformin in Stroke Rodents. Stroke Res Treat 2017; 2017:9756429. [PMID: 28634570 PMCID: PMC5467394 DOI: 10.1155/2017/9756429] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes are major causes of death and disability. Searching for potential therapeutic strategies to prevent and treat stroke is necessary, given the increase in overall life expectancy. Epidemiological reports indicate that metformin is an oral antidiabetic medication that can reduce the incidence of ischemic events in patients with diabetes mellitus. Its mechanism of action has not been elucidated, but metformin pleiotropic effects involve actions in addition to glycemic control. AMPK activation has been described as one of the pharmacological mechanisms that explain the action of metformin and that lead to neuroprotective effects. Most experiments done in the cerebral ischemia model, via middle cerebral artery occlusion in rodents (MCAO), had positive results favoring metformin's neuroprotective role and involve several cellular pathways like oxidative stress, endothelial nitric oxide synthase activation, activation of angiogenesis and neurogenesis, autophagia, and apoptosis. We will review the pharmacological properties of metformin and its possible mechanisms that lead to neuroprotection in cerebral ischemia.
Collapse
Affiliation(s)
| | - Mauricio Palacios
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia
| |
Collapse
|
18
|
Pineda-Ramírez N, Gutiérrez Aguilar GF, Espinoza-Rojo M, Aguilera P. Current evidence for AMPK activation involvement on resveratrol-induced neuroprotection in cerebral ischemia. Nutr Neurosci 2017; 21:229-247. [DOI: 10.1080/1028415x.2017.1284361] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Narayana Pineda-Ramírez
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía ‘Manuel Velasco Suárez’, Ciudad de México, 14269, México
| | - Germán Fernando Gutiérrez Aguilar
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía ‘Manuel Velasco Suárez’, Ciudad de México, 14269, México
| | - Mónica Espinoza-Rojo
- Laboratorio de Biología Molecular y Genómica, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, 39087, México
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía ‘Manuel Velasco Suárez’, Ciudad de México, 14269, México
| |
Collapse
|
19
|
Shen P, Hou S, Zhu M, Zhao M, Ouyang Y, Feng J. Cortical spreading depression preconditioning mediates neuroprotection against ischemic stroke by inducing AMP-activated protein kinase-dependent autophagy in a rat cerebral ischemic/reperfusion injury model. J Neurochem 2017; 140:799-813. [PMID: 27987215 DOI: 10.1111/jnc.13922] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/30/2016] [Accepted: 12/02/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Pingping Shen
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
| | - Shuai Hou
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
| | - Mingqin Zhu
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
| | - Mingming Zhao
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
| | - Yibing Ouyang
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
- Department of Anesthesia; Stanford University School of Medicine; Stanford California USA
| | - Jiachun Feng
- Institute of Neuroscience Center and Neurology Department; The First Affiliated Hospital of Jilin University; Changchun Jilin China
| |
Collapse
|
20
|
Abstract
Ischemic preconditioning (IPC) is a robust neuroprotective phenomenon in which a brief period of cerebral ischemia confers transient tolerance to subsequent ischemic challenge. Research on IPC has implicated cellular, molecular, and systemic elements of the immune response in this phenomenon. Potent molecular mediators of IPC include innate immune signaling pathways such as Toll-like receptors and type 1 interferons. Brain ischemia results in release of pro- and anti-inflammatory cytokines and chemokines that orchestrate the neuroinflammtory response, resolution of inflammation, and transition to neurological recovery and regeneration. Cellular mediators of IPC include microglia, the resident central nervous system immune cells, astrocytes, and neurons. All of these cell types engage in cross-talk with each other using a multitude of signaling pathways that modulate activation/suppression of each of the other cell types in response to ischemia. As the postischemic neuroimmune response evolves over time there is a shift in function toward provision of trophic support and neuroprotection. Peripheral immune cells infiltrate the central nervous system en masse after stroke and are largely detrimental, with a few subtypes having beneficial, protective effects, though the role of these immune cells in IPC is largely unknown. The role of neural progenitor cells in IPC-mediated neuroprotection is another active area of investigation as is the role of microglial proliferation in this setting. A mechanistic understanding of these molecular and cellular mediators of IPC may not only facilitate more effective direct application of IPC to specific clinical scenarios, but also, more broadly, reveal novel targets for therapeutic intervention in stroke.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
21
|
Pre-stroke Metformin Treatment is Neuroprotective Involving AMPK Reduction. Neurochem Res 2016; 41:2719-2727. [PMID: 27350579 DOI: 10.1007/s11064-016-1988-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022]
Abstract
Long-term metformin treatment reduces the risk of stroke. However, the effective administration pattern and indications of metformin on acute cerebral ischemia are unclear. To investigate the neuroprotective treatment duration and dosage of metformin on focal ischemia mice and the association of neuroprotection with 5'-adenosine monophosphate-activated protein kinase (AMPK) regulations, male C57BL/6 mice were subjected to permanent or transient middle cerebral artery occlusion (MCAO) and metformin of 3, 10 and 30 mg/kg was intraperitoneally injected 1, 3 or 7 days prior to MCAO, or at the onset, or 1, 3 or 6 h after reperfusion, respectively. Infarct volumes, neurological deficit score, cell apoptosis, both total and phosphorylated AMPK expressions were assessed. Results showed that prolonged pretreatment to 7 days of metformin (10 mg/kg) significantly ameliorated brain infarct, neurological scores and cell apoptosis in permanent MCAO mice. Shorter (3 days or 1 day) or without pretreatment of metformin was not effective, suggesting a pretreatment time window. In transient MCAO mice, metformin showed no neuroprotection even with pretreatment. The expressions of total and phosphorylated AMPK were sharply decreased with effective metformin pretreatments in ischemic brains. Our data provided the first evidence that in acute ischemic injury, a 7-days pretreatment duration of 10 mg/kg metformin is necessary for its neuroprotection, and metformin may not be beneficial in the cases of blood reperfusion.
Collapse
|
22
|
Repeat remote ischaemic pre-conditioning for improved cardiovascular function in humans: A systematic review. IJC HEART & VASCULATURE 2016; 11:55-58. [PMID: 28616526 PMCID: PMC5441349 DOI: 10.1016/j.ijcha.2016.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/04/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Single exposure to remote ischaemic pre-conditioning (RIPC) has been shown to be effective in reducing major adverse events during cardiac surgery. We evaluated the efficacy of repeated exposure RIPC to elicit improvements in cardiovascular function. METHODS A systematic search was conducted up until May 1st, 2015, using the following databases: EMBASE, PubMed (Medline), Web of Science and the Cochrane Central Registry of Controlled Trials (CENTRAL). Data was extracted and synthesized from published studies of repeat RIPC. RESULTS Data from seven studies showed evidence of improvements in vascular function and anti-hypertensive effects of systolic, diastolic and mean arterial blood pressure following repeat RIPC. Currently existing work justifies a systematic review but not data pooling of individual study data. Repeat RIPC has also produced evidence of improvements in endothelial dependent vasodilation, but not non-endothelial dependent vasodilation, cutaneous vascular conductance or cardiorespiratory fitness. CONCLUSION Repeated RIPC exposure has produced evidence of improvements in endothelial dependent vasodilation, ulcer healing and blood pressure but no benefit in non-endothelial dependent vasodilation, cutaneous vascular conductance or cardiorespiratory fitness. The optimal delivery of RIPC remains unclear, but at least 3 or preferably 4, 5 min exposures appears to be most beneficial, at least for reducing blood pressure. Aside from those undertaking cardiac surgery, other study populations with endothelial dysfunction may benefit from repeat exposure to RIPC.
Collapse
|
23
|
Chen W, Wei S, Yu Y, Xue H, Yao F, Zhang M, Xiao J, Hatch GM, Chen L. Pretreatment of rats with increased bioavailable berberine attenuates cerebral ischemia-reperfusion injury via down regulation of adenosine-5'monophosphate kinase activity. Eur J Pharmacol 2016; 779:80-90. [PMID: 26957053 DOI: 10.1016/j.ejphar.2016.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 01/09/2023]
Abstract
Berberine (BBR) exhibits multiple beneficial biological effects. However, poor bioavailability of BBR has limited its clinical application. We previously demonstrated that solid dispersion of BBR with sodium caprate (HGSD) remarkably improves its bioavailability. We examined whether this increased bioavailability of BBR could protect the brain from ischemia-reperfusion (IR) induced injury. Rats treated with HGSD, SC and saline for 7 days then subjected to cerebral ischemia reperfusion by middle cerebral artery occlusion for 2h followed 12h reperfusion. Neurological deficit scores, infarct size, SOD, MDA and NO levels were examined. P-AMPK, Bax, cleaved-Caspase-3 in brain was determined. To further probe for the mechanism of beneficial effect of HGSD, PC12 cells were incubated with serum from control or HGSD pretreated animals, incubated with 300μM H2O2 to induce apoptosis. Caspase-3 activity and cell apoptosis was evaluated. HGSD pretreatment significantly attenuated neurological deficit scores, reduced infarct size, increased SOD and decreased MDA and NO after cerebral IR injury compared to controls. Meanwhile, HGSD pretreatment significantly reduced expression of p-AMPK, Bax, cleaved-Caspase-3 after cerebral IR injury. Sodium caprate (100mg/kg/d) pretreatment alone did not exhibit any of these beneficial effects. PC12 cell apoptosis was attenuated when cells were cultured with HGSD serum compared to control. The presence of AMPK activator (AICAR) attenuated whereas AMPK inhibitor (Compound C) augmented the protective effect of HGSD serum on PC12 cell apoptosis.The results indicate that HGSD-pretreatment of rats protects the brain from ischemia-reperfusion injury and the mechanism is due to its anti-apoptotic effect mediated by decreased activation of AMPK.
Collapse
Affiliation(s)
- Weijia Chen
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China; Department of Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin, China
| | - Shengnan Wei
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China
| | - Yang Yu
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China
| | - Huan Xue
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China
| | - Fan Yao
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China.
| | - Jun Xiao
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, Jilin, China.
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics, University of Manitoba, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun Jilin, China
| |
Collapse
|
24
|
Systematic review of survival time in experimental mouse stroke with impact on reliability of infarct estimation. J Neurosci Methods 2016; 261:10-8. [PMID: 26620203 DOI: 10.1016/j.jneumeth.2015.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/24/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Stroke is the second most common cause of death worldwide. Only one treatment for acute ischemic stroke is currently available, thrombolysis with rt-PA, but it is limited in its use. Many efforts have been invested in order to find additive treatments, without success. A multitude of reasons for the translational problems from mouse experimental stroke to clinical trials probably exists, including infarct size estimations around the peak time of edema formation. Furthermore, edema is a more prominent feature of stroke in mice than in humans, because of the tendency to produce larger infarcts with more substantial edema. PURPOSE This paper will give an overview of previous studies of experimental mouse stroke, and correlate survival time to peak time of edema formation. Furthermore, investigations of whether the included studies corrected the infarct measurements for edema and a comparison of correction methods will be discussed. METHOD Relevant terms were searched in the National Library of Medicine PubMed database. A method for classification of infarct measurement methods was made using a naming convention. CONCLUSION Our study shows that infarct size estimations are often performed around the peak time of edema, with a median of 24h. Most studies do consider edema formation, however, there is no consensus on what method to use to correct for edema. Furthermore, investigations into neuroprotective drugs should use longer survival times to ensure completion of the investigated process. Our findings indicate a need for more research in this area, and establishment of common correction methodology.
Collapse
|
25
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
26
|
Ran QQ, Chen HL, Liu YL, Yu HX, Shi F, Wang MS. Electroacupuncture preconditioning attenuates ischemic brain injury by activation of the adenosine monophosphate-activated protein kinase signaling pathway. Neural Regen Res 2015; 10:1069-75. [PMID: 26330828 PMCID: PMC4541236 DOI: 10.4103/1673-5374.160095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 01/16/2023] Open
Abstract
Electroacupuncture has therapeutic effects on ischemic brain injury, but its mechanism is still poorly understood. In this study, mice were stimulated by electroacupuncture at the Baihui (GV20) acupoint for 30 minutes at 1 mA and 2/15 Hz for 5 consecutive days. A cerebral ischemia model was established by ligating the bilateral common carotid artery for 15 minutes. At 72 hours after injury, neuronal injury in the mouse hippocampus had lessened, and the number of terminal deoxynucleotide transferase-mediated dUTP nick-end labeling-positive cells reduced after electroacupuncture treatment. Moreover, expression of adenosine monophosphate-activated protein kinase α (AMPKα) and phosphorylated AMPKα was up-regulated. Intraperitoneal injection of the AMPK antagonist, compound C, suppressed this phenomenon. Our findings suggest that electroacupuncture preconditioning alleviates ischemic brain injury via AMPK activation.
Collapse
Affiliation(s)
- Qiang-Qiang Ran
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Huai-Long Chen
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Yan-Li Liu
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Hai-Xia Yu
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Fei Shi
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Ming-Shan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
27
|
Thushara Vijayakumar N, Sangwan A, Sharma B, Majid A, Rajanikant GK. Cerebral Ischemic Preconditioning: the Road So Far…. Mol Neurobiol 2015; 53:2579-93. [PMID: 26081149 DOI: 10.1007/s12035-015-9278-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 06/02/2015] [Indexed: 12/25/2022]
Abstract
Cerebral preconditioning constitutes the brain's adaptation to lethal ischemia when first exposed to mild doses of a subtoxic stressor. The phenomenon of preconditioning has been largely studied in the heart, and data from in vivo and in vitro models from past 2-3 decades have provided sufficient evidence that similar machinery exists in the brain as well. Since preconditioning results in a transient protective phenotype labeled as ischemic tolerance, it can open many doors in the medical warfare against stroke, a debilitating cerebrovascular disorder that kills or cripples thousands of people worldwide every year. Preconditioning can be induced by a variety of stimuli from hypoxia to pharmacological anesthetics, and each, in turn, induces tolerance by activating a multitude of proteins, enzymes, receptors, transcription factors, and other biomolecules eventually leading to genomic reprogramming. The intracellular signaling pathways and molecular cascades behind preconditioning are extensively being investigated, and several first-rate papers have come out in the last few years centered on the topic of cerebral ischemic tolerance. However, translating the experimental knowledge into the clinical scaffold still evades practicality and faces several challenges. Of the various preconditioning strategies, remote ischemic preconditioning and pharmacological preconditioning appears to be more clinically relevant for the management of ischemic stroke. In this review, we discuss current developments in the field of cerebral preconditioning and then examine the potential of various preconditioning agents to confer neuroprotection in the brain.
Collapse
Affiliation(s)
- N Thushara Vijayakumar
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Amit Sangwan
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Bhargy Sharma
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Arshad Majid
- Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - G K Rajanikant
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
28
|
Venna VR, Benashski SE, Chauhan A, McCullough LD. Inhibition of glycogen synthase kinase-3β enhances cognitive recovery after stroke: the role of TAK1. ACTA ACUST UNITED AC 2015; 22:336-43. [PMID: 26077686 PMCID: PMC4478333 DOI: 10.1101/lm.038083.115] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/23/2015] [Indexed: 02/06/2023]
Abstract
Memory deficits are common among stroke survivors. Identifying neuroprotective agents that can prevent memory impairment or improve memory recovery is a vital area of research. Glycogen synthase kinase-3β (GSK-3β) is involved in several essential intracellular signaling pathways. Unlike many other kinases, GSK-3β is active only when dephosphorylated and activation promotes inflammation and apoptosis. In contrast, increased phosphorylation leads to reduced GSK-3β (pGSK-3β) activity. GSK-3β inhibition has beneficial effects on memory in other disease models. GSK-3β regulates both the 5'AMP-activated kinase (AMPK) and transforming growth factor-β-activated kinase (TAK1) pathways. In this work, we examined the effect of GSK-3β inhibition, both independently, in conjunction with a TAK inhibitor, and in AMPK-α2 deficient mice, after stroke to investigate mechanistic interactions between these pathways. GSK-3β inhibition was neuroprotective and ameliorated stroke-induced cognitive impairments. This was independent of AMPK signaling as the protective effects of GSK-3β inhibition were seen in AMPK deficient mice. However, GSK-3β inhibition provided no additive protection in mice treated with a TAK inhibitor suggesting that TAK1 is an upstream regulator of GSK-3β. Targeting GSK-3β could be a novel therapeutic strategy for post-stroke cognitive deficits.
Collapse
Affiliation(s)
- Venugopal Reddy Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | - Sharon E Benashski
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | - Anjali Chauhan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | - Louise D McCullough
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA MC-1840, Department of Neurology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA The Stroke Center at Hartford Hospital, Hartford, Connecticut 06102, USA
| |
Collapse
|
29
|
Li P, Su L, Li X, Di W, Zhang X, Zhang C, He T, Zhu X, Zhang Y, Li Y. Remote limb ischemic postconditioning protects mouse brain against cerebral ischemia/reperfusion injury via upregulating expression of Nrf2, HO-1 and NQO-1 in mice. Int J Neurosci 2015; 126:552-559. [DOI: 10.3109/00207454.2015.1042973] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
30
|
Ma Y, Bu J, Dang H, Sha J, Jing Y, Shan-jiang AI, Li H, Zhu Y. Inhibition of adenosine monophosphate-activated protein kinase reduces glial cell-mediated inflammation and induces the expression of Cx43 in astroglias after cerebral ischemia. Brain Res 2015; 1605:1-11. [DOI: 10.1016/j.brainres.2014.11.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/25/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|
31
|
Rousset CI, Leiper FC, Kichev A, Gressens P, Carling D, Hagberg H, Thornton C. A dual role for AMP-activated protein kinase (AMPK) during neonatal hypoxic-ischaemic brain injury in mice. J Neurochem 2015; 133:242-52. [PMID: 25598140 PMCID: PMC4855681 DOI: 10.1111/jnc.13034] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 02/03/2023]
Abstract
Perinatal hypoxic–ischaemic encephalopathy (HIE) occurs in 1–2 in every 1000 term infants and the devastating consequences range from cerebral palsy, epilepsy and neurological deficit to death. Cellular damage post insult occurs after a delay and is mediated by a secondary neural energy failure. AMP‐activated protein kinase (AMPK) is a sensor of cellular stress resulting from ATP depletion and/or calcium dysregulation, hallmarks of the neuronal cell death observed after HIE. AMPK activation has been implicated in the models of adult ischaemic injury but, as yet, there have been no studies defining its role in neonatal asphyxia. Here, we find that in an in vivo model of neonatal hypoxia–ischaemic and in oxygen/glucose deprivation in neurons, there is pathological activation of the calcium/calmodulin‐dependent protein kinase kinase β (CaMKKβ)‐AMPKα1 signalling pathway. Pharmacological inhibition of AMPK during the insult promotes neuronal survival but, conversely, inhibiting AMPK activity prior to the insult sensitizes neurons, exacerbating cell death. Our data have pathological relevance for neonatal HIE as prior sensitization such as exposure to bacterial infection (reported to reduce AMPK activity) produces a significant increase in injury.
![]() We show that in an in vivo model of neonatal hypoxia–ischaemic and in oxygen/glucose deprivation in neurons, there is a pathological activation of the CaMKKβ‐AMPKα1 signalling pathway. Inhibiting AMPK during OGD promotes neuronal survival; conversely, inhibiting AMPK prior to OGD exacerbates cell death. Our data have clinical relevance as prior sensitization (e.g. exposure to bacterial infection reducing AMPK activity) increases injury. AMPK, AMP‐activated protein kinase; HI, hypoxia–ischaemia; OGD, oxygen–glucose deprivation.
Collapse
Affiliation(s)
- Catherine I Rousset
- Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Zhao Z, Sui Y, Gao W, Cai B, Fan D. Effects of diet on adenosine monophosphate-activated protein kinase activity and disease progression in an amyotrophic lateral sclerosis model. J Int Med Res 2014; 43:67-79. [PMID: 25534414 DOI: 10.1177/0300060514554725] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES To study the effects of diet on disease progression and activity levels of adenosine monophosphate-activated protein kinase (AMPK), and its downstream targets, in an amyotrophic lateral sclerosis (ALS) animal model. METHODS AMPK activity was measured in cerebral cortex, spinal cord, cerebellum and hindlimb muscle tissue using immunohistochemistry in transgenic mice overexpressing human superoxide dismutase-1 (SOD1(G93A)) fed a high-fat (HFD), standard ad libitum (AL) or calorie-restricted (CR) diet; AMPK activity was also measured in wild-type (SOD1(WT)) mice. Activity of AMPK and phospho-AMPK, acetyl coenzyme-A carboxylase (ACC), phospho-ACC and heat shock protein-70 (Hsp70) were also measured using Western blot. Food intake and grip strength were recorded; body composition was analysed using dual energy X-ray absorptiometry. Motor neuron survival was observed using Nissl staining. RESULTS AMPK activity increased and Hsp70 expression decreased in AL SOD1(G93A) mice compared with SOD1(WT) mice in spinal cord and hindlimb muscle. Compared with AL SOD1(G93A) mice, CR SOD1(G93A) mice showed increased AMPK activity, downregulated Hsp70 expression, reduced motor neuron survival in spinal cord and hindlimb muscle and reduced lifespan; HFD SOD1(G93A) mice showed opposite effects. CONCLUSIONS In this mouse model, increased AMPK activity seems to play a negative role in motor neuron survival, possibly through a novel mechanism involving Hsp70 downregulation. These changes can be modified by diet. Inhibition of AMPK may provide a therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Zichun Zhao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Yanling Sui
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Wenchao Gao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Bin Cai
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
33
|
Venna VR, Li J, Hammond MD, Mancini NS, McCullough LD. Chronic metformin treatment improves post-stroke angiogenesis and recovery after experimental stroke. Eur J Neurosci 2014; 39:2129-38. [PMID: 24649970 DOI: 10.1111/ejn.12556] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 02/06/2023]
Abstract
Metformin is currently the first-line treatment drug for type 2 diabetes. Metformin is a well-known activator of AMP-activated protein kinase (AMPK). In experimental studies, metformin has been shown to exert direct vascular effects by increasing vascular endothelial growth factor expression and improving microvascular density. As stroke is the leading cause of long-term disability and angiogenesis is implicated as an important mechanism in functional recovery, we hypothesized that chronic metformin treatment would improve post-stroke functional recovery by enhancing functional microvascular density. For this study, C57BL/6N male mice were subjected to a 60-min middle cerebral artery occlusion, and were given 50 mg/kg/day metformin beginning 24 h post-stroke for 3 weeks. Behavioral recovery was assessed using adhesive-tape removal and the apomorphine-induced turning test. The role of angiogenesis was assessed by counting vessel branch points from fluorescein-conjugated lectin-perfused brain sections. Importantly even if metformin treatment was initiated 24 h after injury it enhanced recovery and significantly improved stroke-induced behavioral deficits. This recovery occurred in parallel with enhanced angiogenesis and with restoration of endogenous cerebral dopaminergic tone and revascularization of ischemic tissue. We assessed if the effects on recovery and angiogenesis were mediated by AMPK. When tested in AMPK α-2 knockout mice, we found that metformin treatment did not have the same beneficial effects on recovery and angiogenesis, suggesting that metformin-induced angiogenic effects are mediated by AMPK. The results from this study suggest that metformin mediates post-stroke recovery by enhancing angiogenesis, and these effects are mediated by AMPK signaling.
Collapse
Affiliation(s)
- Venugopal R Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | |
Collapse
|
34
|
Ashabi G, Khodagholi F, Khalaj L, Goudarzvand M, Nasiri M. Activation of AMP-activated protein kinase by metformin protects against global cerebral ischemia in male rats: interference of AMPK/PGC-1α pathway. Metab Brain Dis 2014; 29:47-58. [PMID: 24435937 DOI: 10.1007/s11011-013-9475-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/16/2013] [Indexed: 12/25/2022]
Abstract
Here, we have investigated the effect of metformin pretreatment in the rat models of global cerebral ischemia. Cerebral ischemia which leads to brain dysfunction is one of the main causes of neurodegeneration and death worldwide. Metformin is used in clinical drug therapy protocols of diabetes. It is suggested that metformin protects cells under hypoxia and ischemia in non-neuronal contexts. Protective effects of metformin may be modulated via activating the AMP activated protein kinase (AMPK). Our results showed that induction of 30 min global cerebral I/R injury using 4-vesseles occlusion model led to significant cell death in the rat brain. Metformin pretreatment (200 mg kg/once/day, p.o., 2 weeks) attenuated apoptotic cell death and induced mitochondrial biogenesis proteins in the ischemic rats, analyzed using histological and Western blot assays. Besides, inhibition of AMPK by compound c showed that metformin resulted in apoptosis attenuation via AMPK activation. Interestingly, AMPK activation was also involved in the induction of mitochondrial biogenesis proteins using metformin, inhibition of AMPK by compound c reversed such effect, further supporting the role of AMPK upstream of mitochondrial biogenesis proteins. In summary, Metformin pretreatment is able to modulate mitochondrial biogenesis and apoptotic cell death pathways through AMPK activation in the context of global cerebral ischemia, conducting the outcome towards neuroprotection.
Collapse
Affiliation(s)
- Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | | | | |
Collapse
|
35
|
Sinnett SE, Brenman JE. Past strategies and future directions for identifying AMP-activated protein kinase (AMPK) modulators. Pharmacol Ther 2014; 143:111-8. [PMID: 24583089 DOI: 10.1016/j.pharmthera.2014.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/13/2014] [Indexed: 12/30/2022]
Abstract
AMP-activated protein kinase (AMPK) is a promising therapeutic target for cancer, type II diabetes, and other illnesses characterized by abnormal energy utilization. During the last decade, numerous labs have published a range of methods for identifying novel AMPK modulators. The current understanding of AMPK structure and regulation, however, has propelled a paradigm shift in which many researchers now consider ADP to be an additional regulatory nucleotide of AMPK. How can the AMPK community apply this new understanding of AMPK signaling to translational research? Recent insights into AMPK structure, regulation, and holoenzyme-sensitive signaling may provide the hindsight needed to clearly evaluate the strengths and weaknesses of past AMPK drug discovery efforts. Improving future strategies for AMPK drug discovery will require pairing the current understanding of AMPK signaling with improved experimental designs.
Collapse
Affiliation(s)
- Sarah E Sinnett
- Neurobiology Curriculum, University of North Carolina at Chapel Hill (UNC), United States
| | - Jay E Brenman
- UNC Neuroscience Center, United States; Department of Cell Biology and Physiology, UNC, United States.
| |
Collapse
|
36
|
Han Z, Cao J, Song D, Tian L, Chen K, Wang Y, Gao L, Yin Z, Fan Y, Wang C. Autophagy is involved in the cardioprotection effect of remote limb ischemic postconditioning on myocardial ischemia/reperfusion injury in normal mice, but not diabetic mice. PLoS One 2014; 9:e86838. [PMID: 24466263 PMCID: PMC3900658 DOI: 10.1371/journal.pone.0086838] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/15/2013] [Indexed: 11/25/2022] Open
Abstract
Background Recent animal study and clinical trial data suggested that remote limb ischemic postconditioning (RIPostC) can invoke potent cardioprotection. However, during ischemia reperfusion injury (IR), the effect and mechanism of RIPostC on myocardium in subjects with or without diabetes mellitus (DM) are poorly understood. Autophagy plays a crucial role in alleviating myocardial IR injury. The aim of this study was to determine the effect of RIPostC on mice myocardial IR injury model with or without DM, and investigate the role of autophagy in this process. Methodology and Results Streptozocin (STZ) induced DM mice model and myocardial IR model were established. Using a noninvasive technique, RIPostC was induced in normal mice (ND) and DM mice by three cycles of ischemia (5 min) and reperfusion (5 min) in the left hindlimb. In ND group, RIPostC significantly reduced infarct size (32.6±3.0% in ND-RIPostC vs. 50.6±2.4% in ND-IR, p<0.05) and improved cardiac ejection fraction (49.70±3.46% in ND-RIPostC vs. 31.30±3.95% in ND-IR, p<0.05). However, in DM group, no RIPostC mediated cardioprotetion effect was observed. To analyze the role of autophagy, western blot and immunohistochemistry was performed. Our data showed that a decreased sequestosome 1 (SQSTM1/p62) level, an increased Beclin-1 level, and higher ratio of LC3-II/LC3-I were observed in ND RIPostC group, but not DM RIPostC group. Conclusions The current study suggested that RIPostC exerts cardioprotection effect on IR in normal mice, but not DM mice, and this difference is via, at least in part, the up-regulation of autophagy.
Collapse
Affiliation(s)
- Zhihua Han
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Jiatian Cao
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Dongqiang Song
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Lei Tian
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Kan Chen
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Yue Wang
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Lin Gao
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Zhaofang Yin
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
| | - Yuqi Fan
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
- * E-mail: (YF); (CW)
| | - Changqian Wang
- Department of Cardiology, Ninth People’s Hospital, Shanghai Jiaotong University Medical School, PR China
- * E-mail: (YF); (CW)
| |
Collapse
|
37
|
Majid A. Neuroprotection in stroke: past, present, and future. ISRN NEUROLOGY 2014; 2014:515716. [PMID: 24579051 PMCID: PMC3918861 DOI: 10.1155/2014/515716] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 09/16/2013] [Indexed: 01/05/2023]
Abstract
Stroke is a devastating medical condition, killing millions of people each year and causing serious injury to many more. Despite advances in treatment, there is still little that can be done to prevent stroke-related brain damage. The concept of neuroprotection is a source of considerable interest in the search for novel therapies that have the potential to preserve brain tissue and improve overall outcome. Key points of intervention have been identified in many of the processes that are the source of damage to the brain after stroke, and numerous treatment strategies designed to exploit them have been developed. In this review, potential targets of neuroprotection in stroke are discussed, as well as the various treatments that have been targeted against them. In addition, a summary of recent progress in clinical trials of neuroprotective agents in stroke is provided.
Collapse
Affiliation(s)
- Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
- Department of Neurology and Manchester Academic Health Sciences Centre, Salford Royal Hospital, Stott Lane, Salford M6 8HD, UK
| |
Collapse
|
38
|
Manwani B, McCullough LD. Function of the master energy regulator adenosine monophosphate-activated protein kinase in stroke. J Neurosci Res 2013; 91:1018-29. [PMID: 23463465 PMCID: PMC4266469 DOI: 10.1002/jnr.23207] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 12/24/2012] [Accepted: 01/05/2013] [Indexed: 01/09/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is an evolutionarily conserved signaling molecule that is emerging as one of the most important energy sensors in the body. AMPK monitors cellular energy status and is activated via phosphorylation when energy stores are low. This allows for maintenance of energy homeostasis by promoting catabolic pathways for ATP production and limiting processes that consume ATP. Growing number of stimuli have been shown to activate AMPK, and AMPK has been implicated in many diverse biological processes, including cell polarity, autophagy, and senescence. The effect of AMPK activation and its biological functions are extremely diverse and depend on both the overall energy "milieu" and the location and duration of activation. AMPK has tissue- and isoform-specific functions in the brain vs. periphery. These functions and the pathways activated also appear to differ by cell location (hypothalamus vs. cortex), cell type (astrocyte vs. neuron), and duration of exposure. Short bursts of AMPK activation have been found to be involved in ischemic preconditioning and neuronal survival; however, prolonged AMPK activity during ischemia leads to neuronal cell death. AMPK may also underlie some of the beneficial effects of hypothermia, a potential therapy for ischemic brain injury. This review discusses the role of AMPK in ischemic stroke, a condition of severe energy depletion.
Collapse
Affiliation(s)
- Bharti Manwani
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | - Louise D. McCullough
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
- Department of Neurology, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
39
|
Jia J, Xiao Y, Wang W, Qing L, Xu Y, Song H, Zhen X, Ao G, Alkayed NJ, Cheng J. Differential mechanisms underlying neuroprotection of hydrogen sulfide donors against oxidative stress. Neurochem Int 2013; 62:1072-8. [PMID: 23587562 DOI: 10.1016/j.neuint.2013.04.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/05/2013] [Accepted: 04/04/2013] [Indexed: 12/27/2022]
Abstract
This study investigated whether slow-releasing organic hydrogen sulfide donors act through the same mechanisms as those of inorganic donors to protect neurons from oxidative stress. By inducing oxidative stress in a neuronal cell line HT22 with glutamate, we investigated the protective mechanisms of the organic donors: ADT-OH [5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione], the most widely used moiety for synthesizing slow-releasing hydrogen sulfide donors, and ADT, a methyl derivative of ADT-OH. The organic donors were more potent than the inorganic donor sodium hydrogensulfide (NaHS) in protecting HT22 cells against glutamate toxicity. Consistent with previous publications, NaHS partially restored glutamate-depleted glutathione (GSH) levels, protected HT22 from direct free radical damage induced by hydrogen peroxide (H2O2), and NaHS protection was abolished by a KATP channel blocker glibenclamide. However, neither ADT nor ADT-OH enhanced glutamate-depleted GSH levels or protected HT22 from H2O2-induced oxidative stress. Glibenclamide, which abolished NaHS neuroprotection against oxidative stress, did not block ADT and ADT-OH neuroprotection against glutamate-induced oxidative stress. Unexpectedly, we found that glutamate induced AMPK activation and that compound C, a well-established AMPK inhibitor, remarkably protected HT22 from glutamate-induced oxidative stress, suggesting that AMPK activation contributed to oxidative glutamate toxicity. Interestingly, all hydrogen sulfide donors, including NaHS, remarkably attenuated glutamate-induced AMPK activation. However, under oxidative glutamate toxicity, compound C only increased the viability of HT22 cells treated with NaHS, but did not further increase ADT and ADT-OH neuroprotection. Thus, suppressing AMPK activation likely contributed to ADT and ADT-OH neuroprotection. In conclusion, hydrogen sulfide donors acted through differential mechanisms to confer neuroprotection against oxidative toxicity and suppressing AMPK activation was a possible mechanism underlying neuroprotection of organic hydrogen sulfide donors against oxidative toxicity.
Collapse
Affiliation(s)
- Jia Jia
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Karuppagounder SS, Basso M, Sleiman SF, Ma TC, Speer RE, Smirnova NA, Gazaryan IG, Ratan RR. In vitro ischemia suppresses hypoxic induction of hypoxia-inducible factor-1α by inhibition of synthesis and not enhanced degradation. J Neurosci Res 2013; 91:1066-75. [PMID: 23456821 DOI: 10.1002/jnr.23204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/04/2012] [Accepted: 12/22/2012] [Indexed: 01/08/2023]
Abstract
Hypoxia-inducible factor (HIF) mediates a broad, conserved adaptive response to hypoxia, and the HIF pathway is a potential therapeutic target in cerebral ischemia. This study investigated the mechanism by which in vitro ischemia (oxygen-glucose deprivation; OGD) affects canonical hypoxic HIF-1α stabilization. We validated the use of a reporter containing the oxygen-dependent degradation domain of HIF-1α fused to firefly luciferase (ODD-luc) to monitor quantitatively distinct biochemical events leading to hypoxic HIF-1α expression or stabilization in a human neuroblastoma cell line (SH-SY5Y). When OGD was imposed following a 2-hr hypoxic stabilization of ODD-luc, the levels of the reporter were reduced, consistent with prior models proposing that OGD enhances HIF prolylhydroxylase (PHD) activity. Surprisingly, PHD inhibitors and proteasome inhibitors do not stabilize ODD-luc in OGD. Furthermore, OGD does not affect the half-life of ODD-luc protein following hypoxia, suggesting that OGD abrogates hypoxic HIF-1α induction by reducing HIF-1α synthesis rather than by enhancing its degradation. We observed ATP depletion under OGD vs. hypoxia and propose that ATP depletion enhances translational suppression, overcoming the selective synthesis of HIF concurrent with global decreases in protein synthesis in hypoxia. Taken together, these findings biochemically characterize a practical reporter for monitoring HIF-1α levels and support a novel model for HIF regulation in an in vitro model of human ischemia.
Collapse
Affiliation(s)
- Saravanan S Karuppagounder
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Venna VR, Weston G, Benashski SE, Tarabishy S, Liu F, Li J, Conti LH, McCullough LD. NF-κB contributes to the detrimental effects of social isolation after experimental stroke. Acta Neuropathol 2012; 124:425-38. [PMID: 22562356 DOI: 10.1007/s00401-012-0990-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/22/2012] [Accepted: 04/22/2012] [Indexed: 01/07/2023]
Abstract
Social isolation (SI) is increasingly recognized as a risk factor for stroke. Individuals with lack of social support systems have an increased incidence of stroke, poorer recovery, and greater functional decline after injury compared to individuals with social support. Attesting to the importance of social factors in stroke outcome is that these same effects can be reproducibly demonstrated in animals; social interaction improves behavioral deficits and reduces damage after experimental stroke, whereas SI enhances injury. The mechanism by which SI exacerbates injury is unclear. We investigated the role of nuclear factor-kappaB (NF-κB) signaling in male mice that were pair housed (PH) with an ovariectomized female prior to random assignment into continued PH or SI for 7 days prior to middle cerebral artery occlusion. The effects of SI on infarct volume and functional recovery were assessed at 72 h post-stroke. Nuclear NF-κB levels and activity were assessed by Western blot and transcriptional assays. SI significantly exacerbated infarct size in both male and female mice compared to PH mice. SI mice had delayed functional recovery compared to PH mice. An elevation of systemic IL-6 levels, increased nuclear NF-κB transcriptional activity, and enhanced nuclear translocation of NF-κB was seen in SI stroke animals. Interference with NF-κB signaling using either a pharmacological inhibitor or genetically engineered NF-κB p50 knockout mice abolished the detrimental effects of SI on both infarct size and functional recovery. This suggests that NF-κB mediates the detrimental effects of SI.
Collapse
Affiliation(s)
- Venugopal Reddy Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | | | |
Collapse
|