151
|
Srivastava IN, Shperdheja J, Baybis M, Ferguson T, Crino PB. mTOR pathway inhibition prevents neuroinflammation and neuronal death in a mouse model of cerebral palsy. Neurobiol Dis 2015; 85:144-154. [PMID: 26459113 DOI: 10.1016/j.nbd.2015.10.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/23/2015] [Accepted: 10/08/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Mammalian target of rapamycin (mTOR) pathway signaling governs cellular responses to hypoxia and inflammation including induction of autophagy and cell survival. Cerebral palsy (CP) is a neurodevelopmental disorder linked to hypoxic and inflammatory brain injury however, a role for mTOR modulation in CP has not been investigated. We hypothesized that mTOR pathway inhibition would diminish inflammation and prevent neuronal death in a mouse model of CP. METHODS Mouse pups (P6) were subjected to hypoxia-ischemia and lipopolysaccharide-induced inflammation (HIL), a model of CP causing neuronal injury within the hippocampus, periventricular white matter, and neocortex. mTOR pathway inhibition was achieved with rapamycin (an mTOR inhibitor; 5mg/kg) or PF-4708671 (an inhibitor of the downstream p70S6kinase, S6K, 75 mg/kg) immediately following HIL, and then for 3 subsequent days. Phospho-activation of the mTOR effectors p70S6kinase and ribosomal S6 protein and expression of hypoxia inducible factor 1 (HIF-1α) were assayed. Neuronal cell death was defined with Fluoro-Jade C (FJC) and autophagy was measured using Beclin-1 and LC3II expression. Iba-1 labeled, activated microglia were quantified. RESULTS Neuronal death, enhanced HIF-1α expression, and numerous Iba-1 labeled, activated microglia were evident at 24 and 48 h following HIL. Basal mTOR signaling, as evidenced by phosphorylated-S6 and -S6K levels, was unchanged by HIL. Rapamycin or PF-4,708,671 treatment significantly reduced mTOR signaling, neuronal death, HIF-1α expression, and microglial activation, coincident with enhanced expression of Beclin-1 and LC3II, markers of autophagy induction. CONCLUSIONS mTOR pathway inhibition prevented neuronal death and diminished neuroinflammation in this model of CP. Persistent mTOR signaling following HIL suggests a failure of autophagy induction, which may contribute to neuronal death in CP. These results suggest that mTOR signaling may be a novel therapeutic target to reduce neuronal cell death in CP.
Collapse
Affiliation(s)
- Isha N Srivastava
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Jona Shperdheja
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Marianna Baybis
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Tanya Ferguson
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Peter B Crino
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, United States.
| |
Collapse
|
152
|
PAK2 is an effector of TSC1/2 signaling independent of mTOR and a potential therapeutic target for Tuberous Sclerosis Complex. Sci Rep 2015; 5:14534. [PMID: 26412398 PMCID: PMC4585940 DOI: 10.1038/srep14534] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/22/2015] [Indexed: 11/22/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by inactivating mutations in either TSC1 or TSC2 and is characterized by uncontrolled mTORC1 activation. Drugs that reduce mTOR activity are only partially successful in the treatment of TSC, suggesting that mTOR-independent pathways play a role in disease development. Here, kinome profiles of wild-type and Tsc2−/− mouse embryonic fibroblasts (MEFs) were generated, revealing a prominent role for PAK2 in signal transduction downstream of TSC1/2. Further investigation showed that the effect of the TSC1/2 complex on PAK2 is mediated through RHEB, but is independent of mTOR and p21RAC. We also demonstrated that PAK2 over-activation is likely responsible for the migratory and cell cycle abnormalities observed in Tsc2−/− MEFs. Finally, we detected high levels of PAK2 activation in giant cells in the brains of TSC patients. These results show that PAK2 is a direct effector of TSC1-TSC2-RHEB signaling and a new target for rational drug therapy in TSC.
Collapse
|
153
|
Nichols M, Zhang J, Polster BM, Elustondo PA, Thirumaran A, Pavlov EV, Robertson GS. Synergistic neuroprotection by epicatechin and quercetin: Activation of convergent mitochondrial signaling pathways. Neuroscience 2015; 308:75-94. [PMID: 26363153 DOI: 10.1016/j.neuroscience.2015.09.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 08/25/2015] [Accepted: 09/03/2015] [Indexed: 01/08/2023]
Abstract
In view of evidence that increased consumption of epicatechin (E) and quercetin (Q) may reduce the risk of stroke, we have measured the effects of combining E and Q on mitochondrial function and neuronal survival following oxygen-glucose deprivation (OGD). Relative to mouse cortical neuron cultures pretreated (24h) with either E or Q (0.1-10μM), E+Q synergistically attenuated OGD-induced neuronal cell death. E, Q and E+Q (0.3μM) increased spare respiratory capacity but only E+Q (0.3μM) preserved this crucial parameter of neuronal mitochondrial function after OGD. These improvements were accompanied by corresponding increases in cyclic AMP response element binding protein (CREB) phosphorylation and the expression of CREB-target genes that promote neuronal survival (Bcl-2) and mitochondrial biogenesis (PGC-1α). Consistent with these findings, E+Q (0.1 and 1.0μM) elevated mitochondrial gene expression (MT-ND2 and MT-ATP6) to a greater extent than E or Q after OGD. Q (0.3-3.0μM), but not E (3.0μM), elevated cytosolic calcium (Ca(2+)) spikes and the mitochondrial membrane potential. Conversely, E and E+Q (0.1 and 0.3μM), but not Q (0.1 and 0.3μM), activated protein kinase B (Akt). Nitric oxide synthase (NOS) inhibition with L-N(G)-nitroarginine methyl ester (1.0μM) blocked neuroprotection by E (0.3μM) or Q (1.0μM). Oral administration of E+Q (75mg/kg; once daily for 5days) reduced hypoxic-ischemic brain injury. These findings suggest E and Q activate Akt- and Ca(2+)-mediated signaling pathways that converge on NOS and CREB resulting in synergistic improvements in neuronal mitochondrial performance which confer profound protection against ischemic injury.
Collapse
Affiliation(s)
- M Nichols
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Sciences Research Institute, 1348 Summer Street, P.O. Box 15000, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - J Zhang
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Sciences Research Institute, 1348 Summer Street, P.O. Box 15000, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - B M Polster
- Department of Anesthesiology, Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - P A Elustondo
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - A Thirumaran
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Sciences Research Institute, 1348 Summer Street, P.O. Box 15000, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - E V Pavlov
- Department of Basic Sciences, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA.
| | - G S Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Department of Psychiatry, 5909 Veterans' Memorial Lane, 8th Floor Abbie J. Lane Memorial Building, QEII Health Sciences Centre, Halifax, Nova Scotia B3H 2E2, Canada.
| |
Collapse
|
154
|
Gao C, Cai Y, Zhang X, Huang H, Wang J, Wang Y, Tong X, Wang J, Wu J. Ischemic Preconditioning Mediates Neuroprotection against Ischemia in Mouse Hippocampal CA1 Neurons by Inducing Autophagy. PLoS One 2015; 10:e0137146. [PMID: 26325184 PMCID: PMC4556686 DOI: 10.1371/journal.pone.0137146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/12/2015] [Indexed: 12/04/2022] Open
Abstract
The hippocampal CA1 region is sensitive to hypoxic and ischemic injury but can be protected by ischemic preconditioning (IPC). However, the mechanism through which IPC protects hippocampal CA1 neurons is still under investigation. Additionally, the role of autophagy in determining the fate of hippocampal neurons is unclear. Here, we examined whether IPC induced autophagy to alleviate hippocampal CA1 neuronal death in vitro and in vivo with oxygen glucose deprivation (OGD) and bilateral carotid artery occlusion (BCCAO) models. Survival of hippocampal neurons increased from 51.5% ± 6.3% in the non-IPC group (55 min of OGD) to 77.3% ± 7.9% in the IPC group (15 min of OGD, followed by 55 min of OGD 24 h later). The number of hippocampal CA1 layer neurons increased from 182 ± 26 cells/mm2 in the non-IPC group (20 min of BCCAO) to 278 ± 55 cells/mm2 in the IPC group (1 min × 3 BCCAO, followed by 20 min of BCCAO 24 h later). Akt phosphorylation and microtubule-associated protein light chain 3 (LC3)-II/LC3-I expression were increased in the preconditioning group. Moreover, the protective effects of IPC were abolished only by inhibiting the activity of autophagy, but not by blocking the activation of Akt in vitro. Using in vivo experiments, we found that LC3 expression was upregulated, accompanied by an increase in neuronal survival in hippocampal CA1 neurons in the preconditioning group. The neuroprotective effects of IPC on hippocampal CA1 neurons were completely inhibited by treatment with 3-MA. In contrast, hippocampal CA3 neurons did not show changes in autophagic activity or beneficial effects of IPC. These data suggested that IPC may attenuate ischemic injury in hippocampal CA1 neurons through induction of Akt-independent autophagy.
Collapse
Affiliation(s)
- Chunlin Gao
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Ying Cai
- Department of Neuroscience, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin, China
| | - Xuebin Zhang
- Department of Pathology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Huiling Huang
- Department of Neuroscience, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin, China
| | - Jin Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Yajing Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Xiaoguang Tong
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin, China
| | - Jinhuan Wang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin, China
| | - Jialing Wu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
- * E-mail:
| |
Collapse
|
155
|
Wang P, Zhang N, Liang J, Li J, Han S, Li J. Micro-RNA-30a regulates ischemia-induced cell death by targeting heat shock protein HSPA5 in primary cultured cortical neurons and mouse brain after stroke. J Neurosci Res 2015; 93:1756-68. [DOI: 10.1002/jnr.23637] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 07/27/2015] [Accepted: 08/06/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Peng Wang
- Department of Neurobiology and Center of Stroke; Beijing Institute for Brain Disorders, Capital Medical University; Beijing People's Republic of China
- Central Laboratory, Liaoning Medical University; Jinzhou People's Republic of China
| | - Nan Zhang
- Department of Anatomy; Capital Medical University; Beijing People's Republic of China
| | - Jia Liang
- Central Laboratory, Liaoning Medical University; Jinzhou People's Republic of China
| | - Jiefei Li
- Department of Neurobiology and Center of Stroke; Beijing Institute for Brain Disorders, Capital Medical University; Beijing People's Republic of China
| | - Song Han
- Department of Neurobiology and Center of Stroke; Beijing Institute for Brain Disorders, Capital Medical University; Beijing People's Republic of China
| | - Junfa Li
- Department of Neurobiology and Center of Stroke; Beijing Institute for Brain Disorders, Capital Medical University; Beijing People's Republic of China
| |
Collapse
|
156
|
Lu Q, Harris VA, Kumar S, Mansour HM, Black SM. Autophagy in neonatal hypoxia ischemic brain is associated with oxidative stress. Redox Biol 2015; 6:516-523. [PMID: 26454246 PMCID: PMC4602363 DOI: 10.1016/j.redox.2015.06.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/25/2015] [Accepted: 06/25/2015] [Indexed: 01/01/2023] Open
Abstract
Autophagy is activated when the neonatal brain exposed to hypoxia ischemia (HI), but the mechanisms underlying its activation and its role in the neuronal cell death associated with HI is unclear. We have previously shown that reactive oxygen species (ROS) derived from nicotinamide adenine dinucleotide phosphate (NADPH) oxidase play an important role in HI-mediated neuronal cell death. Thus, the aim of this study was to determine if ROS is involved in the activation of autophagy in HI-mediated neonatal brain injury and to determine if this is a protective or deleterious pathway. Initial electron microscopy data demonstrated that autophagosome formation is elevated in P7 hippocampal slice cultures exposed to oxygen-glucose deprivation (OGD). This corresponded with increased levels of LC3II mRNA and protein. The autophagy inhibitor, 3-methyladenine (3-MA) effectively reduced LC3II levels and autophagosome formation in hippocampal slice cultures exposed to OGD. Neuronal cell death was significantly attenuated. Finally, we found that the pharmacologic inhibition of NADPH oxidase using apocynin or gp91ds-tat decreased autophagy in hippocampal slice cultures and the rat brain respectively. Thus, our results suggest that an activation of autophagy contributes to neonatal HI brain injury this is oxidative stress dependent.
Collapse
Affiliation(s)
- Qing Lu
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Valerie A Harris
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Sanjv Kumar
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Heidi M Mansour
- Department of Pharmacy Practice & Science, Department of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
157
|
The impact of sleep and hypoxia on the brain: potential mechanisms for the effects of obstructive sleep apnea. Curr Opin Pulm Med 2015; 20:565-71. [PMID: 25188719 DOI: 10.1097/mcp.0000000000000099] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Obstructive sleep apnea (OSA) is a chronic, highly prevalent, multisystem disease, which is still largely underdiagnosed. Its most prominent risk factors, obesity and older age, are on the rise, and its prevalence is expected to grow further. The last few years have seen an exponential increase in studies to determine the impact of OSA on the central nervous system. OSA-induced brain injury is now a recognized clinical entity, although its possible dual relationship with several other neuropsychiatric and neurodegenerative disorders is debated. The putative neuromechanisms behind some of the effects of OSA on the central nervous system are discussed in this review, focusing on the nocturnal intermittent hypoxia and sleep fragmentation. RECENT FINDINGS Recent preclinical and clinical findings suggest that neurogenic ischemic preconditioning occurs in some OSA patients, and that it may partly explain variability in clinical findings to date. However, the distinct parameters of the interplay between ischemic preconditioning, neuroinflammation, sleep fragmentation and cerebrovascular changes in OSA-induced brain injury are still largely unclear, and more research is required. SUMMARY Early diagnosis and intervention in patients with OSA is of paramount importance. Future clinical studies should utilize multimodal investigative approaches to enable more reliable referencing for the acuity of the pathological process, as well as its reversibility following the treatment.
Collapse
|
158
|
Sleep apnoea and the brain: a complex relationship. THE LANCET RESPIRATORY MEDICINE 2015; 3:404-14. [DOI: 10.1016/s2213-2600(15)00090-9] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 01/23/2023]
|
159
|
Fang Y, Tan J, Zhang Q. Signaling pathways and mechanisms of hypoxia-induced autophagy in the animal cells. Cell Biol Int 2015; 39:891-8. [PMID: 25808799 DOI: 10.1002/cbin.10463] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/10/2015] [Indexed: 12/19/2022]
Abstract
Hypoxia occurs in a series of supraphysiological circumstances, for instance, sleep disorders, myocardial infarction and cerebral stroke, that can induce a systematic inflammatory response. Such a response may then lead to a widespread dysfunction and cell injury. Autophagy, a cellular homeostatic process that governs the turnover of damaged organelles and proteins, can be triggered by multiple forms of extra- and intracellular stress, for example, hypoxia, nutrient deprivation and reactive oxygen specie. Central to this process is the formation of double-membrane vesicles, thereby autophagosomes sequester portions of cytosol and deliver them to the lysosomes for a breakdown. In recent years, several distinct oxygen-sensing pathways that regulate the cellular response to autophagy have been defined. For instance, hypoxia influences autophagy in part through the activation of the hypoxia-inducible factor (HIF)-dependent pathways. In chronic and moderate hypoxia, autophagy plays a protective role by mediating the removal of the damaged organelles and protein. Moreover, three additional oxygen-sensitive signaling pathways are also associated with the activation of autophagy. These include mammalian target of rapamycin (mTOR) kinase, unfolded protein response (UPR)- and PKCδ-JNK1-dependent pathways. Contrary to the protective effects of autophagy, during rapid and severe oxygen fluctuations, autophagy may be detrimental and induce cell death. In this review, we highlight a serious of recent advances on how autophagy is regulated at the molecular level and on final consequences of cell under different hypoxic environment.
Collapse
Affiliation(s)
- Yungyun Fang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
160
|
The divergent roles of autophagy in ischemia and preconditioning. Acta Pharmacol Sin 2015; 36:411-20. [PMID: 25832421 PMCID: PMC4387298 DOI: 10.1038/aps.2014.151] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/20/2014] [Indexed: 12/11/2022] Open
Abstract
Autophagy is an evolutionarily conserved and lysosome-dependent process for degrading and recycling cellular constituents. Autophagy is activated following an ischemic insult or preconditioning, but it may exert dual roles in cell death or survival during these two processes. Preconditioning or lethal ischemia may trigger autophagy via multiple signaling pathways involving endoplasmic reticulum (ER) stress, AMPK/TSC/mTOR, Beclin 1/BNIP3/SPK2, and FoxO/NF-κB transcription factors, etc. Autophagy then interacts with apoptotic and necrotic signaling pathways to regulate cell death. Autophagy may also maintain cell function by removing protein aggregates or damaged mitochondria. To date, the dual roles of autophagy in ischemia and preconditioning have not been fully clarified. The purpose of the present review is to summarize the recent progress in the mechanisms underlying autophagy activation during ischemia and preconditioning. A better understanding of the dual effects of autophagy in ischemia and preconditioning could help to develop new strategies for the preventive treatment of ischemia.
Collapse
|
161
|
Autophagy in neuronal cells: general principles and physiological and pathological functions. Acta Neuropathol 2015; 129:337-62. [PMID: 25367385 DOI: 10.1007/s00401-014-1361-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/21/2014] [Accepted: 10/25/2014] [Indexed: 12/12/2022]
Abstract
Autophagy delivers cytoplasmic components and organelles to lysosomes for degradation. This pathway serves to degrade nonfunctional or unnecessary organelles and aggregate-prone and oxidized proteins to produce substrates for energy production and biosynthesis. Macroautophagy delivers large aggregates and whole organelles to lysosomes by first enveloping them into autophagosomes that then fuse with lysosomes. Chaperone-mediated autophagy (CMA) degrades proteins containing the KFERQ-like motif in their amino acid sequence, by transporting them from the cytosol across the lysosomal membrane into the lysosomal lumen. Autophagy is especially important for the survival and homeostasis of postmitotic cells like neurons, because these cells are not able to dilute accumulating detrimental substances and damaged organelles by cell division. Our current knowledge on the autophagic pathways and molecular mechanisms and regulation of autophagy will be summarized in this review. We will describe the physiological functions of macroautophagy and CMA in neuronal cells. Finally, we will summarize the current evidence showing that dysfunction of macroautophagy and/or CMA contributes to neuronal diseases. We will give an overview of our current knowledge on the role of autophagy in aging neurons, and focus on the role of autophagy in four types of neurodegenerative diseases, i.e., amyotrophic lateral sclerosis and frontotemporal dementia, prion diseases, lysosomal storage diseases, and Parkinson's disease.
Collapse
|
162
|
Jiang T, Yu JT, Zhu XC, Wang HF, Tan MS, Cao L, Zhang QQ, Gao L, Shi JQ, Zhang YD, Tan L. Acute metformin preconditioning confers neuroprotection against focal cerebral ischaemia by pre-activation of AMPK-dependent autophagy. Br J Pharmacol 2015; 171:3146-57. [PMID: 24611741 DOI: 10.1111/bph.12655] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 02/09/2014] [Accepted: 02/18/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Recent clinical trials report that metformin, an activator of AMP-activated protein kinase (AMPK) used to treat type 2 diabetes, significantly reduces the risk of stroke by actions that are independent of its glucose-lowering effects. However, the underlying molecular mechanisms are not known. Here, we tested the possibility that acute metformin preconditioning confers neuroprotection by pre-activation of AMPK-dependent autophagy in a rat model of permanent middle cerebral artery occlusion (pMCAO). EXPERIMENTAL APPROACH Male Sprague-Dawley rats were pretreated with either vehicle, an AMPK inhibitor, Compound C, or an autophagy inhibitor, 3-methyladenine, and were injected with a single dose of metformin (10 mg kg(-1), i.p.). Then, AMPK activity and autophagy biomarkers in the brain were assessed. At 24 h after metformin treatment, rats were subjected to pMCAO; infarct volume, neurological deficits and cell apoptosis were evaluated 24 and 96 h later. KEY RESULTS A single dose of metformin significantly activated AMPK and induced autophagy in the brain. The enhanced autophagic activity was inhibited by Compound C pretreatment. Furthermore, acute metformin preconditioning significantly reduced infarct volume, neurological deficits and cell apoptosis during a subsequent focal cerebral ischaemia. The neuroprotection mediated by metformin preconditioning was fully abolished by Compound C and partially inhibited by 3-methyladenine. CONCLUSIONS AND IMPLICATIONS These results provide the first evidence that acute metformin preconditioning induces autophagy by activation of brain AMPK, which confers neuroprotection against subsequent cerebral ischaemia. This suggests that metformin, a well-known hypoglycaemic drug, may have a practical clinical use for stroke prevention.
Collapse
Affiliation(s)
- Teng Jiang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Song L, Su M, Wang S, Zou Y, Wang X, Wang Y, Cui H, Zhao P, Hui R, Wang J. MiR-451 is decreased in hypertrophic cardiomyopathy and regulates autophagy by targeting TSC1. J Cell Mol Med 2014; 18:2266-74. [PMID: 25209900 PMCID: PMC4224559 DOI: 10.1111/jcmm.12380] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/30/2014] [Indexed: 01/15/2023] Open
Abstract
The molecular mechanisms that drive the development of cardiac hypertrophy in hypertrophic cardiomyopathy (HCM) remain elusive. Accumulated evidence suggests that microRNAs are essential regulators of cardiac remodelling. We have been suggested that microRNAs could play a role in the process of HCM. To uncover which microRNAs were changed in their expression, microRNA microarrays were performed on heart tissue from HCM patients (n = 7) and from healthy donors (n = 5). Among the 13 microRNAs that were differentially expressed in HCM, miR-451 was the most down-regulated. Ectopic overexpression of miR-451 in neonatal rat cardiomyocytes (NRCM) decreased the cell size, whereas knockdown of endogenous miR-451 increased the cell surface area. Luciferase reporter assay analyses demonstrated that tuberous sclerosis complex 1 (TSC1) was a direct target of miR-451. Overexpression of miR-451 in both HeLa cells and NRCM suppressed the expression of TSC1. Furthermore, TSC1 was significantly up-regulated in HCM myocardia, which correlated with the decreased levels of miR-451. As TSC1 is a known positive regulator of autophagy, we examined the role of miR-451 in the regulation of autophagy. Overexpression of miR-451 in vitro inhibited the formation of the autophagosome. Conversely, miR-451 knockdown accelerated autophagosome formation. Consistently, an increased number of autophagosomes was observed in HCM myocardia, accompanied by up-regulated autophagy markers, and the lipidated form of LC3 and Beclin-1. Taken together, our findings indicate that miR-451 regulates cardiac hypertrophy and cardiac autophagy by targeting TSC1. The down-regulation of miR-451 may contribute to the development of HCM and may be a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Lei Song
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Harputlugil E, Hine C, Vargas D, Robertson L, Manning BD, Mitchell JR. The TSC complex is required for the benefits of dietary protein restriction on stress resistance in vivo. Cell Rep 2014; 8:1160-70. [PMID: 25131199 PMCID: PMC4260622 DOI: 10.1016/j.celrep.2014.07.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/10/2014] [Accepted: 07/14/2014] [Indexed: 02/07/2023] Open
Abstract
Protein restriction (PR) is important for the benefits of dietary restriction on longevity and stress resistance, but relevant nutrient sensors and downstream effectors in mammals remain poorly defined. We used PR-mediated protection from hepatic ischemia reperfusion injury to probe genetic requirements for the evolutionarily conserved nutrient sensors GCN2 and mTORC1 in stress resistance. One week of PR reduced free amino acids and circulating growth factors, activating GCN2 and mTORC1 repressor tuberous sclerosis complex (TSC). However, although GCN2 was dispensable for PR-induced protection, hepatic TSC1 was required. PR improved hepatic insulin sensitivity in a TSC1-dependent manner prior to ischemia, facilitating increased prosurvival signaling and reduced apoptosis after reperfusion. These benefits were partially abrogated by pharmacological PI3K inhibition or genetic deletion of the insulin receptor in hepatocytes. In conclusion, improved insulin sensitivity upon short-term PR required TSC1, facilitated increased prosurvival signaling after injury, and contributed partially to PR-mediated resistance to clinically relevant ischemia reperfusion injury.
Collapse
Affiliation(s)
- Eylul Harputlugil
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Dorathy Vargas
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Lauren Robertson
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
165
|
Chi W, Meng F, Li Y, Wang Q, Wang G, Han S, Wang P, Li J. Downregulation of miRNA-134 protects neural cells against ischemic injury in N2A cells and mouse brain with ischemic stroke by targeting HSPA12B. Neuroscience 2014; 277:111-22. [PMID: 25003713 DOI: 10.1016/j.neuroscience.2014.06.062] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/20/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
Abstract
MicroRNAs (miRNAs) have emerged as a major regulator in neurological diseases, and understanding their molecular mechanism in modulating cerebral ischemic injury may provide potential therapeutic targets for ischemic stroke. However, as one of 19 differentially expressed miRNAs in mouse brain with middle cerebral artery occlusion (MCAO), the role of miR-134 in ischemic injury is not well understood. In this study, the miR-134 expression level was manipulated both in oxygen-glucose deprivation (OGD)-treated N2A neuroblastoma cells in vitro and mouse brain with MCAO-induced ischemic stroke in vivo, and its possible targets of heat shock protein A5 (HSPA5) and HSPA12B were determined by bioinformatics analysis and dual luciferase assay. The results showed that overexpression of miR-134 exacerbated cell death and apoptosis both in vitro and in vivo. Conversely, downregulating miR-134 levels reduced cell death and apoptosis. Furthermore, non-expression of miR-134 enhanced HSPA12B protein levels in OGD-treated N2A cells as well as in the ischemic region. It could attenuate brain infarction size and neural cell damage, and improve neurological outcomes in mice with ischemic stroke, whereas upregulation of miR-134 had the opposite effect. In addition, HSPA12B was validated to be a target of miR-134 and its short interfering RNAs (siRNAs) could block miR-134 inhibitor-induced neuroprotection in OGD-treated N2A cells. In conclusion, downregulation of miR-134 could induce neuroprotection against ischemic injury in vitro and in vivo by negatively upregulating HSPA12B protein expression.
Collapse
Affiliation(s)
- W Chi
- Department of Anesthesiology, Weifang Medical University, Weifang City 261053, Shangdong Province, PR China
| | - F Meng
- Department of Anesthesiology, Shandong University Affiliated Jinan City Central Hospital, Jinan 250013, PR China.
| | - Y Li
- Department of Anesthesiology, Shandong University Affiliated Jinan City Central Hospital, Jinan 250013, PR China
| | - Q Wang
- Department of Anesthesiology, Shandong University Affiliated Jinan City Central Hospital, Jinan 250013, PR China
| | - G Wang
- Department of Anesthesiology, Weifang Medical University, Weifang City 261053, Shangdong Province, PR China
| | - S Han
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China
| | - P Wang
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China
| | - J Li
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
166
|
Wang P, Xu TY, Wei K, Guan YF, Wang X, Xu H, Su DF, Pei G, Miao CY. ARRB1/β-arrestin-1 mediates neuroprotection through coordination of BECN1-dependent autophagy in cerebral ischemia. Autophagy 2014; 10:1535-48. [PMID: 24988431 PMCID: PMC4206533 DOI: 10.4161/auto.29203] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Autophagy, a highly conserved process conferring cytoprotection against stress, contributes to the progression of cerebral ischemia. β-arrestins are multifunctional proteins that mediate receptor desensitization and serve as important signaling scaffolds involved in numerous physiopathological processes. Here, we show that both ARRB1 (arrestin, β 1) and ARRB2 (arrestin, β 2) were upregulated by cerebral ischemic stress. Knockout of Arrb1, but not Arrb2, aggravated the mortality, brain infarction, and neurological deficit in a mouse model of cerebral ischemia. Accordingly, Arrb1-deficient neurons exhibited enhanced cell injury upon oxygen-glucose deprivation (OGD), an in vitro model of ischemia. Deletion of Arrb1 did not affect the cerebral ischemia-induced inflammation, oxidative stress, and nicotinamide phosphoribosyltransferase upregulation, but markedly suppressed autophagy and induced neuronal apoptosis/necrosis in vivo and in vitro. Additionally, we found that ARRB1 interacted with BECN1/Beclin 1 and PIK3C3/Vps34, 2 major components of the BECN1 autophagic core complex, under the OGD condition but not normal conditions in neurons. Finally, deletion of Arrb1 impaired the interaction between BECN1 and PIK3C3, which is a critical event for autophagosome formation upon ischemic stress, and markedly reduced the kinase activity of PIK3C3. These findings reveal a neuroprotective role for ARRB1, in the context of cerebral ischemia, centered on the regulation of BECN1-dependent autophagosome formation.
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Tian-Ying Xu
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Kai Wei
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Yun-Feng Guan
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Xia Wang
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Hui Xu
- Institute of Biochemistry and Cell Biology; Shanghai Institutes for Biological Sciences; Chinese Academy of Sciences; Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology; Second Military Medical University; Shanghai, China
| | - Gang Pei
- Institute of Biochemistry and Cell Biology; Shanghai Institutes for Biological Sciences; Chinese Academy of Sciences; Shanghai, China; School of Life Science and Technology; Tongji University; Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology; Second Military Medical University; Shanghai, China; Center of Stroke; Beijing Institute for Brain Disorders; Beijing, China
| |
Collapse
|
167
|
Affiliation(s)
- Susan L Stevens
- From the Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland
| | - Keri B Vartanian
- From the Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland
| | - Mary P Stenzel-Poore
- From the Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland.
| |
Collapse
|
168
|
Liu PF, Leung CM, Chang YH, Cheng JS, Chen JJ, Weng CJ, Tsai KW, Hsu CJ, Liu YC, Hsu PC, Pan HW, Shu CW. ATG4B promotes colorectal cancer growth independent of autophagic flux. Autophagy 2014; 10:1454-65. [PMID: 24991826 DOI: 10.4161/auto.29556] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autophagy is reported to suppress tumor proliferation, whereas deficiency of autophagy is associated with tumorigenesis. ATG4B is a deubiquitin-like protease that plays dual roles in the core machinery of autophagy; however, little is known about the role of ATG4B on autophagy and proliferation in tumor cells. In this study, we found that ATG4B knockdown induced autophagic flux and reduced CCND1 expression to inhibit G 1/S phase transition of cell cycle in colorectal cancer cell lines, indicating functional dominance of ATG4B on autophagy inhibition and tumor proliferation in cancer cells. Interestingly, based on the genetic and pharmacological ablation of autophagy, the growth arrest induced by silencing ATG4B was independent of autophagic flux. Moreover, dephosphorylation of MTOR was involved in reduced CCND1 expression and G 1/S phase transition in both cells and xenograft tumors with depletion of ATG4B. Furthermore, ATG4B expression was significantly increased in tumor cells of colorectal cancer patients compared with adjacent normal cells. The elevated expression of ATG4B was highly correlated with CCND1 expression, consistently supporting the notion that ATG4B might contribute to MTOR-CCND1 signaling for G 1/S phase transition in colorectal cancer cells. Thus, we report that ATG4B independently plays a role as a positive regulator on tumor proliferation and a negative regulator on autophagy in colorectal cancer cells. These results suggest that ATG4B is a potential biomarker and drug target for cancer therapy.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan; Department of Biotechnology; Fooyin University; Kaohsiung, Taiwan
| | - Chung-Man Leung
- Department of Radiation Oncology; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan; Department of Safety, Health and Environmental Engineering; National Kaohsiung First University of Science and Technology; Kaohsiung, Taiwan
| | - Yu-Hsiang Chang
- Department of Pediatrics; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan; School of Medicine; National Yang-Ming University; Taipei, Taiwan; Department of Nursing; Tajen University; Pingtung, Taiwan
| | - Jin-Shiung Cheng
- Department of Internal Medicine; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| | - Jih-Jung Chen
- Department of Pharmacy and Graduate Institute of Pharmaceutical Technology; Tajen University; Pingtung, Taiwan
| | - Chung-Jeu Weng
- Department of Obstetrics Gynecology; Zuoying Branch of Kaohsiung Armed Forces General Hospital; Kaohsiung, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| | - Chien-Jen Hsu
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| | - Yen-Chen Liu
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| | - Ping-Chi Hsu
- Department of Safety, Health and Environmental Engineering; National Kaohsiung First University of Science and Technology; Kaohsiung, Taiwan
| | - Hung-Wei Pan
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| | - Chih-Wen Shu
- Department of Medical Education and Research; Kaohsiung Veterans General Hospital; Kaohsiung, Taiwan
| |
Collapse
|
169
|
Wauson EM, Dbouk HA, Ghosh AB, Cobb MH. G protein-coupled receptors and the regulation of autophagy. Trends Endocrinol Metab 2014; 25:274-82. [PMID: 24751357 PMCID: PMC4082244 DOI: 10.1016/j.tem.2014.03.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 01/06/2023]
Abstract
Autophagy is an important catabolic cellular process that eliminates damaged and unnecessary cytoplasmic proteins and organelles. Basal autophagy occurs during normal physiological conditions, but the activity of this process can be significantly altered in human diseases. Thus, defining the regulatory inputs and signals that control autophagy is essential. Nutrients are key modulators of autophagy. Although autophagy is generally accepted to be regulated in a cell-autonomous fashion, recent studies suggest that nutrients can modulate autophagy in a systemic manner by inducing the secretion of hormones and neurotransmitters that regulate G protein-coupled receptors (GPCRs). Emerging studies show that GPCRs also regulate autophagy by directly detecting extracellular nutrients. We review the role of GPCRs in autophagy regulation, highlighting their potential as therapeutic drug targets.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA.
| | - Hashem A Dbouk
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA
| | - Anwesha B Ghosh
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA.
| |
Collapse
|
170
|
Guo W, Feng G, Miao Y, Liu G, Xu C. Rapamycin alleviates brain edema after focal cerebral ischemia reperfusion in rats. Immunopharmacol Immunotoxicol 2014; 36:211-23. [PMID: 24773551 DOI: 10.3109/08923973.2014.913616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Brain edema is a major consequence of cerebral ischemia reperfusion. However, few effective therapeutic options are available for retarding the brain edema progression after cerebral ischemia. Recently, rapamycin has been shown to produce neuroprotective effects in rats after cerebral ischemia reperfusion. Whether rapamycin could alleviate this brain edema injury is still unclear. In this study, the rat stroke model was induced by a 1-h left transient middle cerebral artery occlusion using an intraluminal filament, followed by 48 h of reperfusion. The effects of rapamycin (250 μg/kg body weight, intraperitoneal; i.p.) on brain edema progression were evaluated. The results showed that rapamycin treatment significantly reduced the infarct volume, the water content of the brain tissue and the Evans blue extravasation through the blood-brain barrier (BBB). Rapamycin treatment could improve histological appearance of the brain tissue, increased the capillary lumen space and maintain the integrity of BBB. Rapamycin also inhibited matrix metalloproteinase 9 (MMP9) and aquaporin 4 (AQP4) expression. These data imply that rapamycin could improve brain edema progression after reperfusion injury through maintaining BBB integrity and inhibiting MMP9 and AQP4 expression. The data of this study provide a new possible approach for improving brain edema after cerebral ischemia reperfusion by administration of rapamycin.
Collapse
Affiliation(s)
- Wei Guo
- Department of Neurology, Binzhou Medical College Affiliated Hospital , Binzhou, Shandong Province , China and
| | | | | | | | | |
Collapse
|
171
|
Down-Regulation of miRNA-30a Alleviates Cerebral Ischemic Injury Through Enhancing Beclin 1-Mediated Autophagy. Neurochem Res 2014; 39:1279-91. [DOI: 10.1007/s11064-014-1310-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/16/2014] [Accepted: 04/11/2014] [Indexed: 12/22/2022]
|
172
|
Ginet V, Spiehlmann A, Rummel C, Rudinskiy N, Grishchuk Y, Luthi-Carter R, Clarke PGH, Truttmann AC, Puyal J. Involvement of autophagy in hypoxic-excitotoxic neuronal death. Autophagy 2014; 10:846-60. [PMID: 24674959 DOI: 10.4161/auto.28264] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neuronal autophagy is increased in numerous excitotoxic conditions including neonatal cerebral hypoxia-ischemia (HI). However, the role of this HI-induced autophagy remains unclear. To clarify this role we established an in vitro model of excitotoxicity combining kainate treatment (Ka, 30 µM) with hypoxia (Hx, 6% oxygen) in primary neuron cultures. KaHx rapidly induced excitotoxic death that was completely prevented by MK801 or EGTA. KaHx also stimulated neuronal autophagic flux as shown by a rise in autophagosome number (increased levels of LC3-II and punctate LC3 labeling) accompanied by increases in lysosomal abundance and activity (increased SQSTM1/p62 degradation, and increased LC3-II levels in the presence of lysosomal inhibitors) and fusion (shown using an RFP-GFP-LC3 reporter). To determine the role of the enhanced autophagy we applied either pharmacological autophagy inhibitors (3-methyladenine or pepstatinA/E64) or lentiviral vectors delivering shRNAs targeting Becn1 or Atg7. Both strategies reduced KaHx-induced neuronal death. A prodeath role of autophagy was also confirmed by the enhanced toxicity of KaHx in cultures overexpressing BECN1 or ATG7. Finally, in vivo inhibition of autophagy by intrastriatal injection of a lentiviral vector expressing a Becn1-targeting shRNA increased the volume of intact striatum in a rat model of severe neonatal cerebral HI. These results clearly show a death-mediating role of autophagy in hypoxic-excitotoxic conditions and suggest that inhibition of autophagy should be considered as a neuroprotective strategy in HI brain injuries.
Collapse
Affiliation(s)
- Vanessa Ginet
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Amélie Spiehlmann
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Coralie Rummel
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Nikita Rudinskiy
- Brain Mind Institute; École Polytechnique Fédérale de Lausanne; Lausanne, Switzerland
| | - Yulia Grishchuk
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Ruth Luthi-Carter
- Brain Mind Institute; École Polytechnique Fédérale de Lausanne; Lausanne, Switzerland
| | - Peter G H Clarke
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland
| | - Anita C Truttmann
- Clinic of Neonatology; Department of Pediatrics and Pediatric Surgery; Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences; Faculty of Biology and Medicine; University of Lausanne; Lausanne, Switzerland; Clinic of Neonatology; Department of Pediatrics and Pediatric Surgery; Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| |
Collapse
|
173
|
Di Nardo A, Wertz MH, Kwiatkowski E, Tsai PT, Leech JD, Greene-Colozzi E, Goto J, Dilsiz P, Talos DM, Clish CB, Kwiatkowski DJ, Sahin M. Neuronal Tsc1/2 complex controls autophagy through AMPK-dependent regulation of ULK1. Hum Mol Genet 2014; 23:3865-74. [PMID: 24599401 DOI: 10.1093/hmg/ddu101] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a disorder arising from mutation in the TSC1 or TSC2 gene, characterized by the development of hamartomas in various organs and neurological manifestations including epilepsy, intellectual disability and autism. TSC1/2 protein complex negatively regulates the mammalian target of rapamycin complex 1 (mTORC1) a master regulator of protein synthesis, cell growth and autophagy. Autophagy is a cellular quality-control process that sequesters cytosolic material in double membrane vesicles called autophagosomes and degrades it in autolysosomes. Previous studies in dividing cells have shown that mTORC1 blocks autophagy through inhibition of Unc-51-like-kinase1/2 (ULK1/2). Despite the fact that autophagy plays critical roles in neuronal homeostasis, little is known on the regulation of autophagy in neurons. Here we show that unlike in non-neuronal cells, Tsc2-deficient neurons have increased autolysosome accumulation and autophagic flux despite mTORC1-dependent inhibition of ULK1. Our data demonstrate that loss of Tsc2 results in autophagic activity via AMPK-dependent activation of ULK1. Thus, in Tsc2-knockdown neurons AMPK activation is the dominant regulator of autophagy. Notably, increased AMPK activity and autophagy activation are also found in the brains of Tsc1-conditional mouse models and in cortical tubers resected from TSC patients. Together, our findings indicate that neuronal Tsc1/2 complex activity is required for the coordinated regulation of autophagy by AMPK. By uncovering the autophagy dysfunction associated with Tsc2 loss in neurons, our work sheds light on a previously uncharacterized cellular mechanism that contributes to altered neuronal homeostasis in TSC disease.
Collapse
Affiliation(s)
- Alessia Di Nardo
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Mary H Wertz
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Erica Kwiatkowski
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Peter T Tsai
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Jarrett D Leech
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Emily Greene-Colozzi
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - June Goto
- Division of Translational Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pelin Dilsiz
- Department of Neurology, New York University School of Medicine, New York, NY 10016, USA and
| | - Delia M Talos
- Department of Neurology, New York University School of Medicine, New York, NY 10016, USA and
| | - Clary B Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - David J Kwiatkowski
- Division of Translational Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mustafa Sahin
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA,
| |
Collapse
|
174
|
Ren C, Guingab-Cagmat J, Kobeissy F, Zoltewicz S, Mondello S, Gao M, Hafeez A, Li N, Geng X, Larner SF, Anagli J, Hayes RL, Ji X, Ding Y. A neuroproteomic and systems biology analysis of rat brain post intracerebral hemorrhagic stroke. Brain Res Bull 2014; 102:46-56. [PMID: 24583080 DOI: 10.1016/j.brainresbull.2014.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/19/2014] [Accepted: 02/18/2014] [Indexed: 12/26/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke leading to a high rate of death and disability worldwide. Although it has been hypothesized that much of the IHC insult occurs in the subacute period mediated via a series of complex pathophysiological cascades, the molecular mechanisms involved in ICH have not been systematically characterized. Among the best approaches to understand the underlying mechanisms of injury and recovery, protein dynamics assessment via proteomics/systems biology platforms represent one of the cardinal techniques optimized for mechanisms investigation and biomarker identification. A proteomics approach may provide a biomarker focused framework from which to identify candidate biomarkers of pathophysiological processes involved in brain injury after stroke. In this work, a neuroproteomic approach (LC-MS/MS) was applied to investigate altered expression of proteins that are induced in brain tissue 3 h after injury in a rat model of ICH. Data from sham and focal ischemic models were also obtained and used for comparison. Based on the differentially expressed protein profile, systems biology analysis was conducted to identify associated cellular processes and related interaction maps. After LC-MS/MS analysis of the 3 h brain lysates, 86 proteins were differentially expressed between hemorrhagic and sham tissues. Furthermore, 38 proteins were differentially expressed between ischemic and sham tissues. On the level of global pathway analysis, hemorrhagic stroke proteins were shown to be involved in autophagy, ischemia, necrosis, apoptosis, calpain activation, and cytokine secretion. Moreover, ischemic stroke proteins were related to cell death, ischemia, inflammation, oxidative stress, caspase activation and apoptotic injury. In conclusion, the proteomic responses identified in this study provide key information about target proteins involved in specific pathological pathways.
Collapse
Affiliation(s)
- Changhong Ren
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | - Joy Guingab-Cagmat
- Banyan Labs, Banyan Biomarkers Inc., Alachua, FL, USA; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | - Firas Kobeissy
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida, Gainesville, FL, USA
| | - Susie Zoltewicz
- Banyan Labs, Banyan Biomarkers Inc., Alachua, FL, USA; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | | | - Mingqing Gao
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Adam Hafeez
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ning Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | - Xiaokun Geng
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Stephen F Larner
- Banyan Labs, Banyan Biomarkers Inc., Alachua, FL, USA; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | - John Anagli
- Banyan Labs, Banyan Biomarkers Inc., Alachua, FL, USA
| | - Ronald L Hayes
- Banyan Labs, Banyan Biomarkers Inc., Alachua, FL, USA; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China
| | - Xunming Ji
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Yuchuan Ding
- Xuanwu-Banyan Biomarker Research and Assay Center, Beijing 100053, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
175
|
Wang DB, Uo T, Kinoshita C, Sopher BL, Lee RJ, Murphy SP, Kinoshita Y, Garden GA, Wang HG, Morrison RS. Bax interacting factor-1 promotes survival and mitochondrial elongation in neurons. J Neurosci 2014; 34:2674-83. [PMID: 24523556 PMCID: PMC3921432 DOI: 10.1523/jneurosci.4074-13.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/16/2013] [Accepted: 01/11/2014] [Indexed: 01/04/2023] Open
Abstract
Bax-interacting factor 1 (Bif-1, also known as endophilin B1) is a multifunctional protein involved in the regulation of apoptosis, mitochondrial morphology, and autophagy. Previous studies in non-neuronal cells have shown that Bif-1 is proapoptotic and promotes mitochondrial fragmentation. However, the role of Bif-1 in postmitotic neurons has not been investigated. In contrast to non-neuronal cells, we now report that in neurons Bif-1 promotes viability and mitochondrial elongation. In mouse primary cortical neurons, Bif-1 knockdown exacerbated apoptosis induced by the DNA-damaging agent camptothecin. Neurons from Bif-1-deficient mice contained fragmented mitochondria and Bif-1 knockdown in wild-type neurons also resulted in fragmented mitochondria which were more depolarized, suggesting mitochondrial dysfunction. During ischemic stroke, Bif-1 expression was downregulated in the penumbra of wild-type mice. Consistent with Bif-1 being required for neuronal viability, Bif-1-deficient mice developed larger infarcts and an exaggerated astrogliosis response following ischemic stroke. Together, these data suggest that, in contrast to non-neuronal cells, Bif-1 is essential for the maintenance of mitochondrial morphology and function in neurons, and that loss of Bif-1 renders neurons more susceptible to apoptotic stress. These unique actions may relate to the presence of longer, neuron-specific Bif-1 isoforms, because only these forms of Bif-1 were able to rescue deficiencies caused by Bif-1 suppression. This finding not only demonstrates an unexpected role for Bif-1 in the nervous system but this work also establishes Bif-1 as a potential therapeutic target for the treatment of neurological diseases, especially degenerative disorders characterized by alterations in mitochondrial dynamics.
Collapse
Affiliation(s)
| | - Takuma Uo
- Departments of Neurological Surgery and
| | | | - Bryce L. Sopher
- Neurology, University of Washington School of Medicine, Seattle, Washington 98195-6470, and
| | | | | | | | - Gwenn A. Garden
- Neurology, University of Washington School of Medicine, Seattle, Washington 98195-6470, and
| | - Hong-Gang Wang
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | | |
Collapse
|
176
|
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
|
177
|
Abstract
The four-vessel occlusion (4-VO) method of global forebrain cerebral ischemia mimics the human clinical condition of cardiac arrest. It results in selective neuronal damage and is a useful experimental system to dissect underlying mechanisms behind ischemic phenomena such as the differential susceptibility of CA1 compared to the CA3 region of the hippocampus. It also provides a "proof-of-principle" system for testing out potential agents for neuroprotection.
Collapse
Affiliation(s)
- Gina Hadley
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
178
|
Evodiamine Induces Transient Receptor Potential Vanilloid-1-Mediated Protective Autophagy in U87-MG Astrocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:354840. [PMID: 24454492 PMCID: PMC3884692 DOI: 10.1155/2013/354840] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/23/2013] [Indexed: 12/19/2022]
Abstract
Cerebral ischemia is a leading cause of mortality and morbidity worldwide, which results in cognitive and motor dysfunction, neurodegenerative diseases, and death. Evodiamine (Evo) is extracted from Evodia rutaecarpa Bentham, a plant widely used in Chinese herbal medicine, which possesses variable biological abilities, such as anticancer, anti-inflammation, antiobesity, anti-Alzheimer's disease, antimetastatic, antianoxic, and antinociceptive functions. But the effect of Evo on ischemic stroke is unclear. Increasing data suggest that activation of autophagy, an adaptive response to environmental stresses, could protect neurons from ischemia-induced cell death. In this study, we found that Evo induced autophagy in U87-MG astrocytes. A scavenger of extracellular calcium and an antagonist of transient receptor potential vanilloid-1 (TRPV-1) decreased the percentage of autophagy accompanied by an increase in apoptosis, suggesting that Evo may induce calcium-mediated protective autophagy resulting from an influx of extracellular calcium. The same phenomena were also confirmed by a small interfering RNA technique to knock down the expression of TRPV1. Finally, Evo-induced c-Jun N-terminal kinases (JNK) activation was reduced by a TRPV1 antagonist, indicating that Evo-induced autophagy may occur through a calcium/c-Jun N-terminal kinase (JNK) pathway. Collectively, Evo induced an influx of extracellular calcium, which led to JNK-mediated protective autophagy, and this provides a new option for ischemic stroke treatment.
Collapse
|
179
|
Rosenzweig I, Kempton MJ, Crum WR, Glasser M, Milosevic M, Beniczky S, Corfield DR, Williams SC, Morrell MJ. Hippocampal hypertrophy and sleep apnea: a role for the ischemic preconditioning? PLoS One 2013; 8:e83173. [PMID: 24349453 PMCID: PMC3862721 DOI: 10.1371/journal.pone.0083173] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 10/30/2013] [Indexed: 11/25/2022] Open
Abstract
The full impact of multisystem disease such as obstructive sleep apnoea (OSA) on regions of the central nervous system is debated, as the subsequent neurocognitive sequelae are unclear. Several preclinical studies suggest that its purported major culprits, intermittent hypoxia and sleep fragmentation, can differentially affect adult hippocampal neurogenesis. Although the prospective biphasic nature of chronic intermittent hypoxia in animal models of OSA has been acknowledged, so far the evidence for increased ‘compensatory’ neurogenesis in humans is uncertain. In a cross-sectional study of 32 patients with mixed severity OSA and 32 non-apnoeic matched controls inferential analysis showed bilateral enlargement of hippocampi in the OSA group. Conversely, a trend for smaller thalami in the OSA group was noted. Furthermore, aberrant connectivity between the hippocampus and the cerebellum in the OSA group was also suggested by the correlation analysis. The role for the ischemia/hypoxia preconditioning in the neuropathology of OSA is herein indicated, with possible further reaching clinical implications.
Collapse
Affiliation(s)
- Ivana Rosenzweig
- Department of Neuroimaging, Institute of Psychiatry, King's College, London, United Kingdom
- Danish Epilepsy Centre, Dianalund, Denmark
- * E-mail:
| | - Matthew J. Kempton
- Department of Neuroimaging, Institute of Psychiatry, King's College, London, United Kingdom
| | - William R. Crum
- Department of Neuroimaging, Institute of Psychiatry, King's College, London, United Kingdom
| | - Martin Glasser
- Academic Unit of Sleep and Breathing, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- NIHR Respiratory Disease Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Milan Milosevic
- Department for Environmental and Occupational Health, University of Zagreb, School of Medicine, Andrija Štampar School of Public Health, Zagreb, Croatia
| | - Sandor Beniczky
- Danish Epilepsy Centre, Dianalund, Denmark
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Douglas R. Corfield
- Manchester Medical School, University of Manchester, Manchester, United Kingdom
| | - Steven C. Williams
- Department of Neuroimaging, Institute of Psychiatry, King's College, London, United Kingdom
| | - Mary J. Morrell
- Academic Unit of Sleep and Breathing, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- NIHR Respiratory Disease Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| |
Collapse
|
180
|
Luo T, Park Y, Sun X, Liu C, Hu B. Protein misfolding, aggregation, and autophagy after brain ischemia. Transl Stroke Res 2013; 4:581-8. [PMID: 24323413 DOI: 10.1007/s12975-013-0299-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/17/2013] [Accepted: 10/20/2013] [Indexed: 01/02/2023]
Abstract
Ischemic brain injury is a common disorder linked to a variety of diseases. Significant progress has been made in our understanding of the underlying mechanisms. Previous studies show that protein misfolding, aggregation, and multiple organelle damage are major pathological events in postischemic neurons. The autophagy pathway is the chief route for bulk degradation of protein aggregates and damaged organelles. The latest studies suggest that impairment of autophagy contributes to abnormal protein aggregation and organelle damages after brain ischemia. This article reviews recent studies of protein misfolding, aggregation, and impairment of autophagy after brain ischemia.
Collapse
Affiliation(s)
- Tianfei Luo
- Shock, Trauma and Anesthesiology Research Center, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | | | | | | | | |
Collapse
|
181
|
Nelson MP, Shacka JJ. Autophagy Modulation in Disease Therapy: Where Do We Stand? CURRENT PATHOBIOLOGY REPORTS 2013; 1:239-245. [PMID: 24470989 DOI: 10.1007/s40139-013-0032-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Since it was first described more than 50 years ago autophagy has been examined in many contexts, from cell survival to pathogen sequestration and removal. In more recent years our understanding of autophagy has developed sufficiently to allow effective targeted therapeutics to be developed against various diseases. The field of autophagy research is expanding rapidly, demonstrated by increases in both numbers of investigators in the field and the breadth of topics being addressed. Some diseases, such as the many cancers, have come to the fore in autophagy therapeutics research as a better understanding of their underlying mechanisms has surfaced. Numerous treatments are being developed and explored, from creative applications of the classic autophagy modulators chloroquine and rapamycin, to repurposing drugs approved for other treatments, such as astemizole, which is currently in use as an antimalarial and chronic rhinitis treatment. The landscape of autophagy modulation in disease therapy is rapidly changing and this review hopes to provide a cross-section of the current state of the field.
Collapse
Affiliation(s)
- Michael P Nelson
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| | - John J Shacka
- Department of Pathology, Neuropathology Division, University of Alabama at Birmingham, Birmingham VA Medical Center, Sparks Clinics Room SC 930B, 1720 7 Ave S., Birmingham, AL 35294, USA
| |
Collapse
|
182
|
Hadley G, De Luca GC, Papadakis M, Buchan AM. Endogenous neuroprotection: hamartin modulates an austere approach to staying alive in a recession. Int J Stroke 2013; 8:449-50. [PMID: 23879750 DOI: 10.1111/ijs.12130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tuberous sclerosis complex 1 (hamartin) is an effective endogenous neuroprotectant. Understanding the endogenous mechanism for neuroprotection mediated by hamartin may afford a novel approach to effective treatment of neurological diseases such as stroke, neurodegenerative diseases, and epilepsy, with possible applications to nonneurological conditions.
Collapse
Affiliation(s)
- Gina Hadley
- Acute Stroke Programme, Radcliffe Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
183
|
Crystal structure of the yeast TSC1 core domain and implications for tuberous sclerosis pathological mutations. Nat Commun 2013; 4:2135. [DOI: 10.1038/ncomms3135] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/12/2013] [Indexed: 12/12/2022] Open
|