1
|
Yadava S, Reddy DH, Nakka VP, Anusha VL, Dumala N, Viswanadh MK, Chakravarthi G, Nalluri BN, Ramakrishna K. Unravelling neuroregenerative and neuroprotective roles of Wnt/β-catenin pathway in ischemic stroke: Insights into molecular mechanisms. Neuroscience 2025; 565:527-547. [PMID: 39681254 DOI: 10.1016/j.neuroscience.2024.12.024] [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: 11/01/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Stroke is a serious condition often resulting in mortality or long-term disability, causing cognitive, memory, and motor impairments. A reduction in cerebral blood flow below critical levels defines the ischemic core and penumbra: the core undergoes irreversible damage, while the penumbra remains viable but functionally impaired. This functional impairment activates complex cell signaling pathways that determine cell survival or death, making the penumbra a key target for therapeutic interventions to prevent further damage. The Wnt/β-catenin (WβC) signaling pathway has emerged as a potential neuroprotective mechanism, promoting neurogenesis, angiogenesis, neuronal connectivity, and maintaining blood-brain barrier integrity after stroke. Activation of the WβC pathway also mitigates oxidative stress, inflammation, and apoptosis in ischemic regions, enhancing its neuroprotective effects. However, the overexpression of GSK3β and DKK1, or the presence of their agonists, can counteract these benefits. This review explores the therapeutic potential of WβC signaling, highlighting the effects of pharmacological modulation through antagonists, agonists, synthetic chemicals, natural products, stem cells, and macromolecules in preclinical models of ischemic stroke. While preclinical evidence supports the benefits of WβC activation, its role in human stroke requires further investigation. Additionally, the review discusses the potential adverse effects of prolonged WβC activation and suggests strategies to mitigate them. Overall, WβC signaling holds promise as a therapeutic target, offering insights into stroke pathophysiology and informing the development of novel treatment strategies.
Collapse
Affiliation(s)
- Srikanth Yadava
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | | | - Venkata Prasuja Nakka
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, 500046, India.
| | | | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | - Matte Kasi Viswanadh
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| | | | - Buchi N Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India
| | - Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, India.
| |
Collapse
|
2
|
Zhou R, Hu W, Ma PX, Liu CJ. Versatility of 14-3-3 proteins and their roles in bone and joint-related diseases. Bone Res 2024; 12:58. [PMID: 39406741 PMCID: PMC11480210 DOI: 10.1038/s41413-024-00370-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/30/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Bone and joint-related diseases, including osteoarthritis (OA), rheumatoid arthritis (RA), and bone tumors, pose significant health challenges due to their debilitating effects on the musculoskeletal system. 14-3-3 proteins, a family of conserved regulatory molecules, play a critical role in the pathology of these diseases. This review discusses the intricate structure and multifunctionality of 14-3-3 proteins, their regulation of signaling pathways, and their interactions with other proteins. We underscore the significance of 14-3-3 proteins in the regulation of osteoblasts, osteoclasts, chondrocytes, and bone remodeling, all key factors in the maintenance and dysfunction of bone and joint systems. Specific focus is directed toward elucidating the contribution of 14-3-3 proteins in the pathology of OA, RA, and bone malignancies, where dysregulated 14-3-3-mediated signaling cascades have been implicated in the disease processes. This review illuminates how the perturbation of 14-3-3 protein interactions can lead to the pathological manifestations observed in these disorders, including joint destruction and osteolytic activity. We highlight cutting-edge research that positions 14-3-3 proteins as potential biomarkers for disease progression and as innovative therapeutic targets, offering new avenues for disease intervention and management.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Weirong Hu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Peter X Ma
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Guo T, Chen M, Liu J, Wei Z, Yuan J, Wu W, Wu Z, Lai Y, Zhao Z, Chen H, Liu N. Neuropilin-1 promotes mitochondrial structural repair and functional recovery in rats with cerebral ischemia. J Transl Med 2023; 21:297. [PMID: 37138283 PMCID: PMC10155168 DOI: 10.1186/s12967-023-04125-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
OBJECTIVES Available literature documents that ischemic stroke can disrupt the morphology and function of mitochondria and that the latter in other disease models can be preserved by neuropilin-1 (NRP-1) via oxidative stress suppression. However, whether NRP-1 can repair mitochondrial structure and promote functional recovery after cerebral ischemia is still unknown. This study tackled this very issue and explored the underlying mechanism. METHODS Adeno-associated viral (AAV)-NRP-1 was stereotaxically inoculated into the cortex and ipsilateral striatum posterior of adult male Sprague-Dawley (SD) rats before a 90-min transient middle cerebral artery occlusion (tMCAO) and subsequent reperfusion. Lentivirus (LV)-NRP-1 was transfected into rat primary cortical neuronal cultures before a 2-h oxygen-glucose deprivation and reoxygenation (OGD/R) injury to neurons. The expression and function of NRP-1 and its specific protective mechanism were investigated by Western Blot, immunofluorescence staining, flow cytometry, magnetic resonance imaging, transmission electron microscopy, etc. The binding was detected by molecular docking and molecular dynamics simulation. RESULTS Both in vitro and in vivo models of cerebral ischemia/reperfusion (I/R) injury presented a sharp increase in NRP-1 expression. The expression of AAV-NRP-1 markedly ameliorated the cerebral I/R-induced damage to the motor function and restored the mitochondrial morphology. The expression of LV-NRP-1 alleviated mitochondrial oxidative stress and bioenergetic deficits. AAV-NRP-1 and LV-NRP-1 treatments increased the wingless integration (Wnt)-associated signals and β-catenin nuclear localization. The protective effects of NRP-1 were reversed by the administration of XAV-939. CONCLUSIONS NRP-1 can produce neuroprotective effects against I/R injury to the brain by activating the Wnt/β-catenin signaling pathway and promoting mitochondrial structural repair and functional recovery, which may serve as a promising candidate target in treating ischemic stroke.
Collapse
Affiliation(s)
- Ting Guo
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zengyu Wei
- Emergency Department, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinjin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Yongxing Lai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Hsieh CH, Chou CC, Fang YC, Hsu PH, Chiu YH, Yang CS, Jow GM, Tang CY, Jeng CJ. 14-3-3 proteins regulate cullin 7-mediated Eag1 degradation. Cell Biosci 2023; 13:18. [PMID: 36717938 PMCID: PMC9885684 DOI: 10.1186/s13578-023-00969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Mutations in the human gene encoding the neuron-specific Eag1 (KV10.1; KCNH1) potassium channel are linked to congenital neurodevelopmental diseases. Disease-causing mutant Eag1 channels manifest aberrant gating function and defective protein homeostasis. Both the E3 ubiquitin ligase cullin 7 (Cul7) and the small acid protein 14-3-3 serve as binding partners of Eag1. Cul7 mediates proteasomal and lysosomal degradation of Eag1 protein, whereas over-expression of 14-3-3 notably reduces Eag1 channel activity. It remains unclear whether 14-3-3 may also contribute to Eag1 protein homeostasis. RESULTS In human cell line and native rat neurons, disruptions of endogenous 14-3-3 function with the peptide inhibitor difopein or specific RNA interference up-regulated Eag1 protein level in a transcription-independent manner. Difopein hindered Eag1 protein ubiquitination at the endoplasmic reticulum and the plasma membrane, effectively promoting the stability of both immature and mature Eag1 proteins. Suppression of endogenous 14-3-3 function also reduced excitotoxicity-associated Eag1 degradation in neurons. Difopein diminished Cul7-mediated Eag1 degradation, and Cul7 knock-down abolished the effect of difopein on Eag1. Inhibition of endogenous 14-3-3 function substantially perturbed the interaction of Eag1 with Cul7. Further structural analyses suggested that the intracellular Per-Arnt-Sim (PAS) domain and cyclic nucleotide-binding homology domain (CNBHD) of Eag1 are essential for the regulatory effect of 14-3-3 proteins. Significantly, suppression of endogenous 14-3-3 function reduced Cul7-mediated degradation of disease-associated Eag1 mutant proteins. CONCLUSION Overall these results highlight a chaperone-like role of endogenous 14-3-3 proteins in regulating Eag1 protein homeostasis, as well as a therapeutic potential of 14-3-3 modulators in correcting defective protein expression of disease-causing Eag1 mutants.
Collapse
Affiliation(s)
- Chang-Heng Hsieh
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan
| | - Chia-Cheng Chou
- grid.36020.370000 0000 8889 3720National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Ya-Ching Fang
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.19188.390000 0004 0546 0241Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100 Taiwan
| | - Po-Hao Hsu
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.19188.390000 0004 0546 0241Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100 Taiwan
| | - Yi-Hung Chiu
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan
| | - Chi-Sheng Yang
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan
| | - Guey-Mei Jow
- grid.256105.50000 0004 1937 1063School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Yung Tang
- grid.19188.390000 0004 0546 0241Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100 Taiwan
| | - Chung-Jiuan Jeng
- grid.260539.b0000 0001 2059 7017Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.260539.b0000 0001 2059 7017Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
5
|
Grisley ED, Huber KN, Knapp AN, Butteiger DN, Banz WJ, MacLean JA, Wallace DG, Cheatwood JL. Effects of Dietary Soy Protein Isolate Versus Isoflavones Alone on Poststroke Skilled Ladder Rung Walking and Cortical mRNA Expression Differ in Adult Male Rats. J Med Food 2022; 25:158-165. [PMID: 34936814 PMCID: PMC8867101 DOI: 10.1089/jmf.2020.0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Dietary soy protein isolate (SPI) and the isoflavones daidzein and genistein have been shown to provide neuroprotection from stroke. However, the mechanisms remain uncertain. We sought to determine whether the addition of isoflavones to a diet containing caseinate (CAS) as the protein source would induce behavioral neuroprotection similar to that seen previously in rats fed SPI. Furthermore, we aimed to characterize the baseline and poststroke expression of mRNAs involved in pathways previously published as perhaps mediating soy-based neuroprotection from stroke and other markers of neuronal plasticity, oxidative stress, and inflammation. Adult male rats were fed a semipurified diet containing (1) sodium caseinate (CAS), (2) CAS plus daidzein and genistein (CAS+ISO), or (3) SPI for 2 weeks. A subset of rats was euthanized, and tissue was collected for quantitative real-time PCR (qPCR). Remaining rats underwent a middle cerebral artery occlusion to induce a stroke. Samples for qPCR were collected on day 3 poststroke. Rats fed SPI made fewer errors on the skilled ladder rung walking task after stroke compared to rats fed CAS (P < .05). Rats fed CAS+ISO were not different from rats fed CAS or SPI. Significant effects of diet were found at day 0 for Syp, Pparg, and Ywhae and at day 3 for Rtn4 expression. We concluded that the benefits of SPI are not solely attributable to daidzein and genistein.
Collapse
Affiliation(s)
- Elizabeth Dawn Grisley
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Kalene N. Huber
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Austen N. Knapp
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | | | - William J. Banz
- Department of Animal Science, Food, and Nutrition, Southern Illinois University, Carbondale, Illinois, USA
| | - James A. MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Douglas G. Wallace
- Department of Psychology, Northern Illinois University, Dekalb, Illinois, USA
| | - Joseph L. Cheatwood
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, Illinois, USA.,Address correspondence to: Joseph L. Cheatwood, PhD, Department of Anatomy, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA,
| |
Collapse
|
6
|
Zhou X, Wang Z, Xu B, Ji N, Meng P, Gu L, Li Y. Long non-coding RNA NORAD protects against cerebral ischemia/reperfusion injury induced brain damage, cell apoptosis, oxidative stress and inflammation by regulating miR-30a-5p/YWHAG. Bioengineered 2021; 12:9174-9188. [PMID: 34709972 PMCID: PMC8810080 DOI: 10.1080/21655979.2021.1995115] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
LncRNAs are identified as critical regulators in cerebral ischemia/reperfusion injury (CIRI). In this current work, SH-SY5Y cells suffered from oxygen-glucose deprivation/reperfusion (OGD/R) were applied to analyze the biological role of lncRNA NORAD and underlying molecular mechanism in CIRI in vitro. Levels of lncRNA NORAD, miR-30a-5p and YWHAG were measured using RT-qPCR. Bioinformatics analysis predicted the binding sites of lncRNA NORAD to miR-30a-5p and miR-30a-5p to YWHAG. Luciferase reporter assay verified the binding relationships among lncRNA NORAD, miR-30a-5p and YWHAG. Additionally, cell viability was determined using CCK-8 assay, and cell apoptosis was assessed using TUNEL staining and western blot analysis. Moreover, the levels of ROS, MDA, LDH and SOD as well as IL-1β, TNF-α, and IL-6 were assessed via application of the corresponding assay kits. Decreased cell viability and temporarily increased lncRNA NORAD level were observed in SH-SY5Y cells after OGD/R. It was demonstrated that lncRNA NORAD regulated YWHAG expression by sponging miR-30a-5p. Upregulation of lncRNA NORAD contributed to the enhancement of cell viability, the inhibition of cell apoptosis as well as the alleviation of oxidative stress and inflammation in OGD/R-injured SH-SY5Y cells, which were reversed upon elevation of miR-30a-5p. In contrast, downregulation of lncRNA NORAD reduced cell viability, promoted cell apoptosis as well as aggravated oxidative stress and inflammation under OGD/R challenge, and the functions of lncRNA NORAD knockdown in OGD/R injury were abolished by upregulation of YWHAG. Taken together, lncRNA NORAD exerted protective effects against OGD/R-induced neural injury by sponging miR-30a-5p to upregulate YWHAG expression.
Collapse
Affiliation(s)
- Xinyu Zhou
- Department of Neurology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, the First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Zhonglong Wang
- Department of Neurology, Jining Psychiatric Hospital, Jining, Shandong Province, China
| | - Bingchao Xu
- Department of Neurology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, the First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Niu Ji
- Department of Neurology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, the First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Pin Meng
- Department of Neurology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, the First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Lei Gu
- Rehabilitation Center, Beijing Xiaotangshan Hospital, Beijing, China
| | - Ying Li
- Rehabilitation Center, Beijing Xiaotangshan Hospital, Beijing, China
| |
Collapse
|
7
|
Gómora-García JC, Gerónimo-Olvera C, Pérez-Martínez X, Massieu L. IRE1α RIDD activity induced under ER stress drives neuronal death by the degradation of 14-3-3 θ mRNA in cortical neurons during glucose deprivation. Cell Death Discov 2021; 7:131. [PMID: 34083523 PMCID: PMC8175356 DOI: 10.1038/s41420-021-00518-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/23/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Altered protein homeostasis is associated with neurodegenerative diseases and acute brain injury induced under energy depletion conditions such as ischemia. The accumulation of damaged or unfolded proteins triggers the unfolded protein response (UPR), which can act as a homeostatic response or lead to cell death. However, the factors involved in turning and adaptive response into a cell death mechanism are still not well understood. Several mechanisms leading to brain injury induced by severe hypoglycemia have been described but the contribution of the UPR has been poorly studied. Cell responses triggered during both the hypoglycemia and the glucose reinfusion periods can contribute to neuronal death. Therefore, we have investigated the activation dynamics of the PERK and the IRE1α branches of the UPR and their contribution to neuronal death in a model of glucose deprivation (GD) and glucose reintroduction (GR) in cortical neurons. Results show a rapid activation of the PERK/p-eIF2α/ATF4 pathway leading to protein synthesis inhibition during GD, which contributes to neuronal adaptation, however, sustained blockade of protein synthesis during GR promotes neuronal death. On the other hand, IRE1α activation occurs early during GD due to its interaction with BAK/BAX, while ASK1 is recruited to IRE1α activation complex during GR promoting the nuclear translocation of JNK and the upregulation of Chop. Most importantly, results show that IRE1α RNase activity towards its splicing target Xbp1 mRNA occurs late after GR, precluding a homeostatic role. Instead, IRE1α activity during GR drives neuronal death by positively regulating ASK1/JNK activity through the degradation of 14-3-3 θ mRNA, a negative regulator of ASK and an adaptor protein highly expressed in brain, implicated in neuroprotection. Collectively, results describe a novel regulatory mechanism of cell death in neurons, triggered by the downregulation of 14-3-3 θ mRNA induced by the IRE1α branch of the UPR.
Collapse
Affiliation(s)
- Juan Carlos Gómora-García
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, México
| | - Cristian Gerónimo-Olvera
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, México.,Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Xochitl Pérez-Martínez
- Departamento de Genética Molecular, División de Investigación Básica, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, México
| | - Lourdes Massieu
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, México.
| |
Collapse
|
8
|
Wu P, Zhang B, Han X, Sun Y, Sun Z, Li L, Zhou X, Jin Q, Fu P, Xu W, Qian H. HucMSC exosome-delivered 14-3-3ζ alleviates ultraviolet radiation-induced photodamage via SIRT1 pathway modulation. Aging (Albany NY) 2021; 13:11542-11563. [PMID: 33882455 PMCID: PMC8109102 DOI: 10.18632/aging.202851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/01/2021] [Indexed: 01/05/2023]
Abstract
Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-ex) are nano-sized membrane-bound vesicles that have been reported to facilitate skin regeneration and repair. However, the roles played by hucMSC-ex in ultraviolet (UV) radiation-induced skin photodamage and the underlying mechanisms remain unknown. To investigate the functions of hucMSC-ex in a rat model of acute skin photodamage, immunofluorescence and immunohistochemical staining, quantitative real-time-polymerase chain reaction (qRT-PCR), western blot, and gene silencing assays were performed. We found that the in vivo subcutaneous injection of hucMSC-ex elicited antioxidant and anti-inflammatory effects against UV radiation-induced DNA damage and apoptosis. Further studies showed that the sirtuin 1 (SIRT1) expression level in skin keratinocytes (HaCaT) decreased in a time- and dose-dependent manner under in vitro UV radiation induced-oxidative stress conditions, which could be reversed by treatment with hucMSC-ex. The activation of SIRT1 significantly attenuated UV- and H2O2-induced cytotoxic damage by inhibiting oxidative stress and promoting the activation of autophagy. Our study found that 14-3-3ζ protein, which was delivered by hucMSC-ex, exerted a cytoprotective function via the modulation of a SIRT1-dependent antioxidant pathway. Collectively, our findings indicated that hucMSC-ex might represent a new potential agent for preventing or treating UV radiation-induced skin photodamage and aging.
Collapse
Affiliation(s)
- Peipei Wu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Bin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, Shandong, People's Republic of China
| | - Xinye Han
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Yaoxiang Sun
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Zixuan Sun
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Linli Li
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Xinru Zhou
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Qian Jin
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Peiwen Fu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.,Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| |
Collapse
|
9
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
10
|
Kang JB, Lee SY, Park DJ, Koh PO. Decrease of 14-3-3 proteins by glutamate exposure in the cerebral cortex of newborn rats. Lab Anim Res 2020; 36:8. [PMID: 32257920 PMCID: PMC7119159 DOI: 10.1186/s42826-020-00041-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/12/2020] [Indexed: 11/10/2022] Open
Abstract
Glutamate is a representative excitatory neurotransmitter. However, excessive glutamate exposure causes neuronal cell damage by generating neuronal excitotoxicity. Excitotoxicity in neonates caused by glutamate treatment induces neurological deficits in adults. The 14-3-3 family proteins are conserved proteins that are expressed ubiquitously in a variety of tissues. These proteins contribute to cellular processes, including signal transduction, protein synthesis, and cell cycle control. We proposed that glutamate induces neuronal cell damage by regulating 14-3-3 protein expression in newborn animals. In this study, we investigated the histopathological changes and 14-3-3 proteins expressions as a result of glutamate exposure in the neonatal cerebral cortex. Rat pups at post-natal day 7 were intraperitoneally administrated with vehicle or glutamate (10 mg/kg). Animals were sacrificed 4 h after treatment, and brain tissues were fixed for histological study. Cerebral cortices were isolated and frozen for proteomic study. We observed serious histopathological damages including shrunken dendrites and atypical neurons in glutamate-treated cerebral cortices. In addition, we identified that 14-3-3 family proteins decreased in glutamate-exposed cerebral cortices using a proteomic approach. Moreover, Western blot analysis provided results that glutamate treatment in neonates decreased 14-3-3 family proteins expressions, including the β/α, ζ/δ, γ, ε, τ, and η isoforms. 14-3-3 proteins are involved in signal transduction, metabolism, and anti-apoptotic functions. Thus, our findings suggest that glutamate induces neonatal neuronal cell damage by modulating 14-3-3 protein expression.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Seung-Yun Lee
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| |
Collapse
|
11
|
Kim DE, Cho CH, Sim KM, Kwon O, Hwang EM, Kim HW, Park JY. 14-3-3γ Haploinsufficient Mice Display Hyperactive and Stress-sensitive Behaviors. Exp Neurobiol 2019; 28:43-53. [PMID: 30853823 PMCID: PMC6401549 DOI: 10.5607/en.2019.28.1.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 02/03/2023] Open
Abstract
14-3-3γ plays diverse roles in different aspects of cellular processes. Especially in the brain where 14-3-3γ is enriched, it has been reported to be involved in neurological and psychiatric diseases (e.g. Williams-Beuren syndrome and Creutzfeldt-Jakob disease). However, behavioral abnormalities related to 14-3-3γ deficiency are largely unknown. Here, by using 14-3-3γ deficient mice, we found that homozygous knockout mice were prenatally lethal, and heterozygous mice showed developmental delay relative to wild-type littermate mice. In addition, in behavioral analyses, we found that 14-3-3γ heterozygote mice display hyperactive and depressive-like behavior along with more sensitive responses to acute stress than littermate control mice. These results suggest that 14-3-3γ levels may be involved in the developmental manifestation of related neuropsychiatric diseases. In addition, 14-3-3γ heterozygote mice may be a potential model to study the molecular pathophysiology of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Do Eon Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Chang-Hoon Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Kyoung Mi Sim
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Osung Kwon
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hyung-Wook Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
12
|
Selective 14-3-3γ Upregulation Promotes Beclin-1-LC3-Autophagic Influx via β-Catenin Interaction in Starved Neurons In Vitro and In Vivo. Neurochem Res 2019; 44:849-858. [DOI: 10.1007/s11064-019-02717-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/02/2019] [Indexed: 12/30/2022]
|
13
|
Otsuka S, Sakakima H, Terashi T, Takada S, Nakanishi K, Kikuchi K. Preconditioning exercise reduces brain damage and neuronal apoptosis through enhanced endogenous 14-3-3γ after focal brain ischemia in rats. Brain Struct Funct 2018; 224:727-738. [PMID: 30478609 DOI: 10.1007/s00429-018-1800-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/21/2018] [Indexed: 11/24/2022]
Abstract
14-3-3γ is an important early ischemia-inducible protective factor against ischemic cell death in cerebral cortical neurons. We investigated the anti-apoptosis mechanism of enhanced 14-3-3γ mediated by preconditioning exercise-induced brain ischemic tolerance after stroke. Rats were assigned to four groups: exercise and ischemia (Ex group), ischemia and no exercise (No-Ex group), exercise and no ischemia (Ex-only group), and no exercise and ischemia (control group). Rats were trained on a treadmill for 5 days a week for 3 weeks (running speed, 25 m/min; running duration, 30 min/day). After the exercise program, stroke was induced by left middle cerebral artery occlusion. The infarct volume, neurological deficits, and motor function, as well as expression levels of hypoxia-induced factor-1α (HIF-1α), 14-3-3γ, P2X7 receptors, p-β-catenin Ser37, Bax, and caspase 3 were evaluated by immunohistochemistry and western blotting. The expression of HIF-1α and 14-3-3γ significantly increased in neurons and astrocytes in the Ex-only group. HIF-1α was co-expressed with P2X7 receptor- and GFAP-positive astrocytes. After stroke, the Ex group had significantly reduced brain infarction. HIF-1α and 14-3-3γ significantly increased in the Ex group compared to the No-Ex group. In addition, p-β-catenin Ser37 significantly increased following elevated 14-3-3γ; in contrast, Bax and caspase 3 were significantly reduced in the Ex group. Our findings suggest that preconditioning exercise prior to ischemia induces neuron- and astrocyte-mediated brain ischemic tolerance through increased expression of HIF-1α and 14-3-3γ, which are intrinsic protective factors; the upregulated 14-3-3γ induced by preconditioning exercise reduces ischemic neuronal cell death through the 14-3-3γ/p-β-catenin Ser37/Bax/caspase 3 anti-apoptotic pathway.
Collapse
Affiliation(s)
- Shotaro Otsuka
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Harutoshi Sakakima
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Takuto Terashi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Seiya Takada
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kazuki Nakanishi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kiyoshi Kikuchi
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Japan.
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan.
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan.
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
14
|
Kahl A, Blanco I, Jackman K, Baskar J, Milaganur Mohan H, Rodney-Sandy R, Zhang S, Iadecola C, Hochrainer K. Cerebral ischemia induces the aggregation of proteins linked to neurodegenerative diseases. Sci Rep 2018; 8:2701. [PMID: 29426953 PMCID: PMC5807442 DOI: 10.1038/s41598-018-21063-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
Protein aggregation critically affects cell viability in neurodegenerative diseases, but whether this also occurs in ischemic brain injury remains elusive. Prior studies report the post-ischemic aggregation of ubiquitin, small ubiquitin-related modifier (SUMO) and ribosomes, however whether other proteins are also affected is unknown. Here we employed a proteomic approach to identify the insoluble, aggregated proteome after cerebral ischemia. Mice underwent transient middle cerebral artery occlusion or sham-surgery. After 1-hour reperfusion, prior to apparent brain injury, mice were sacrificed and detergent-insoluble proteins were obtained and identified by nanoLC-MS/MS. Naturally existing insoluble proteins were determined in sham controls and aggregated proteins after cerebral ischemia/reperfusion were identified. Selected aggregated proteins found by proteomics were biochemically verified and aggregation propensities were studied during ischemia with or without reperfusion. We found that ischemia/reperfusion induces the aggregation of RNA-binding and heat-shock proteins, ubiquitin, SUMO and other proteins involved in cell signalling. RNA-binding proteins constitute the largest group of aggregating proteins in ischemia. These include TDP43, FUS, hnRNPA1, PSF/SFPQ and p54/NONO, all of which have been linked to neurodegeneration associated with amyotrophic lateral sclerosis and frontotemporal dementia. The aggregation of neurodegeneration-related disease proteins in cerebral ischemia unveils a previously unappreciated molecular overlap between neurodegenerative diseases and ischemic stroke.
Collapse
Affiliation(s)
- Anja Kahl
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Ismary Blanco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Katherine Jackman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Juhi Baskar
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Harihar Milaganur Mohan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Reunet Rodney-Sandy
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Sheng Zhang
- Institute of Biotechnology and Life Sciences Biotechnologies, Cornell University, Ithaca, NY14853, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065, USA.
| |
Collapse
|
15
|
Liu D, Qiu XY, Wu X, Hu DX, Li CY, Yu SB, Pan F, Chen XQ. Piperlongumine suppresses bladder cancer invasion via inhibiting epithelial mesenchymal transition and F-actin reorganization. Biochem Biophys Res Commun 2017; 494:165-172. [PMID: 29037814 DOI: 10.1016/j.bbrc.2017.10.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022]
Abstract
Piperlongumine (PL), a natural alkaloid isolated from longer pepper plants, is recently found to be a potent selective anti-cancer compound. We first tested its anti-cancer effects on bladder cancer, the fifth most common and aggressive cancer worldwide, to further explore the therapeutic spectrum and molecular mechanisms of PL. PL significantly suppressed bladder cancer cell proliferation, the transition of G2/M phase to next phase, migration/invasion in vitro and bladder cancer growth/development in vivo. PL markedly elevated reactive oxygen species (ROS) and the administration of antioxidants abolished PL induced cell proliferation inhibition, G2/M phase arrest and migration suppression on bladder cancer cells. In vivo studies demonstrated that PL inhibited epithelial mesenchymal transition with profoundly decreased level of Slug, β-catenin, ZEB1 and N-Cadherin. Further, we first reported PL effects on cytoskeleton with prominently reduced lamellipodia formation and decreased F-actin intensity in bladder cancer cells. Taken together, our results first revealed that PL suppressed bladder cancer proliferation and migration in vivo and in vitro, suggesting novel mechanism underlying PL's anti-cancer effect and providing a new anticancer drug strategy for bladder cancer therapy.
Collapse
Affiliation(s)
- Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Yao Qiu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dian Xing Hu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chun Yang Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shang Bin Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Pan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiao Qian Chen
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
16
|
Wang JC, Bindokas VP, Skinner M, Emrick T, Marks JD. Mitochondrial mechanisms of neuronal rescue by F-68, a hydrophilic Pluronic block co-polymer, following acute substrate deprivation. Neurochem Int 2017; 109:126-140. [PMID: 28433663 PMCID: PMC5641222 DOI: 10.1016/j.neuint.2017.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 04/10/2017] [Indexed: 01/09/2023]
Abstract
Global brain ischemia can lead to widespread neuronal death and poor neurologic outcomes in patients. Despite detailed understanding of the cellular and molecular mechanisms mediating neuronal death following focal and global brain hypoxia-ischemia, treatments to reduce ischemia-induced brain injury remain elusive. One pathway central to neuronal death following global brain ischemia is mitochondrial dysfunction, one consequence of which is the cascade of intracellular events leading to mitochondrial outer membrane permeabilization. A novel approach to rescuing injured neurons from death involves targeting cellular membranes using a class of synthetic molecules called Pluronics. Pluronics are triblock copolymers of hydrophilic poly[ethylene oxide] (PEO) and hydrophobic poly[propylene oxide] (PPO). Evidence is accumulating to suggest that hydrophilic Pluronics rescue injured neurons from death following substrate deprivation by preventing mitochondrial dysfunction. Here, we will review current understanding of the nature of interaction of Pluronic molecules with biological membranes and the efficacy of F-68, an 80% hydrophilic Pluronic, in rescuing neurons from injury. We will review data indicating that F-68 reduces mitochondrial dysfunction and mitochondria-dependent death pathways in a model of neuronal injury in vitro, and present new evidence that F-68 acts directly on mitochondria to inhibit mitochondrial outer membrane permeabilization. Finally, we will present results of a pilot, proof-of-principle study suggesting that F-68 is effective in reducing hippocampal injury induced by transient global ischemia in vivo. By targeting mitochondrial dysfunction, F-68 and other Pluronic molecules constitute an exciting new approach to rescuing neurons from acute injury.
Collapse
Affiliation(s)
- Janice C Wang
- Department of Pediatrics, University of Chicago, Chicago, IL, United States
| | - Vytautas P Bindokas
- Department of Pharmacological, Physiological Sciences, University of Chicago, IL, United States
| | - Matthew Skinner
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA, United States
| | - Todd Emrick
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA, United States
| | - Jeremy D Marks
- Department of Pediatrics, University of Chicago, Chicago, IL, United States; Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
17
|
14-3-3 Isoforms Differentially Regulate NFκB Signaling in the Brain After Ischemia-Reperfusion. Neurochem Res 2017; 42:2354-2362. [PMID: 28424948 DOI: 10.1007/s11064-017-2255-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/10/2017] [Accepted: 03/30/2017] [Indexed: 12/21/2022]
Abstract
Mammalian 14-3-3 isoforms exist predominantly in the brain and are heavily involved in neurological diseases. However, the isoform-specific role of 14-3-3 proteins in the brain remains largely unclear. Here, we investigated the role of 14-3-3 isoforms in rat brains after transient middle cerebral artery occlusion and reperfusion. 14-3-3β, η, γ and ζ but not ε or τ were selectively upregulated in cerebral cortical neurons after ischemia-reperfusion (I/R). Selectively, 14-3-3β, γ and ζ were translocated from cytoplasm into the nuclei of neurons after I/R. 14-3-3 bound to p65 and suppressed p65 expression in N2a cells. In the brain, 14-3-3 could either colocalize with p65 in the nuclei of neurons or segregate from p65 expression in cortical neurons after I/R. All evidence together suggests that 14-3-3 isoforms are differentially induced to enter into the nuclei of neurons after I/R, which might regulate NFκB signaling directly or indirectly. Since 14-3-3 proteins are essential for cell survival and NFκB is a key transcriptional factor, our data suggest that the 14-3-3/p65 signaling pathway might be a potential therapeutic target for stroke.
Collapse
|
18
|
Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-Dependent Deacetylase SIRT2 Protects Mouse Brain Against Ischemic Stroke. Mol Neurobiol 2016; 54:7251-7261. [DOI: 10.1007/s12035-016-0173-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/28/2016] [Indexed: 12/20/2022]
|
19
|
Jeon SJ, Sung JH, Koh PO. Hyperglycemia decreases expression of 14-3-3 proteins in an animal model of stroke. Neurosci Lett 2016; 626:13-8. [PMID: 27177727 DOI: 10.1016/j.neulet.2016.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 01/05/2023]
Abstract
Diabetes is a severe metabolic disorder and a major risk factor for stroke. Stroke severity is worse in patients with diabetes compared to the non-diabetic population. The 14-3-3 proteins are a family of conserved acidic proteins that are ubiquitously expressed in cells and tissues. These proteins are involved in many cellular processes including metabolic pathways, signal transduction, protein trafficking, protein synthesis, and cell cycle control. This study investigated 14-3-3 proteins expression in the cerebral cortex of animals with diabetes, cerebral ischemic injury and a combination of both diabetes and cerebral ischemic injury. Diabetes was induced by intraperitoneal injection of streptozotocin (40mg/kg) in adult male rats. After 4 weeks of treatment, middle cerebral artery occlusion (MCAO) was performed for the induction of focal cerebral ischemia and cerebral cortex tissue was collected 24h after MCAO. We confirmed that diabetes increases infarct volume following MCAO compared to non-diabetic animals. In diabetic animals with MCAO injury, reduction of 14-3-3 β/α, 14-3-3 ζ/δ, 14-3-3 γ, and 14-3-3 ε isoforms was detected. The expression of these proteins was significantly decreased in diabetic animals with MCAO injury compared to diabetic-only and MCAO-only animals. Moreover, Western blot analysis ascertained the decreased expression of 14-3-3 family proteins in diabetic animals with MCAO injury, including β/α, ζ/δ, γ, ε, τ, and η isoforms. These results show the changes of 14-3-3 proteins expression in streptozotocin-induced diabetic animals with MCAO injury. Thus, these findings suggest that decreases in 14-3-3 proteins might be involved in the regulation of 14-3-3 proteins under the presence of diabetes following MCAO.
Collapse
Affiliation(s)
- Seong-Jun Jeon
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju 660-701, South Korea
| | - Jin-Hee Sung
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju 660-701, South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju 660-701, South Korea.
| |
Collapse
|
20
|
Gilmartin DJ, Soon A, Thrasivoulou C, Phillips ARJ, Jayasinghe SN, Becker DL. Sustained Release of Cx43 Antisense Oligodeoxynucleotides from Coated Collagen Scaffolds Promotes Wound Healing. Adv Healthc Mater 2016; 5:1786-99. [PMID: 27253638 DOI: 10.1002/adhm.201600175] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/21/2016] [Indexed: 12/26/2022]
Abstract
Antisense oligodeoxynucleotides targeting the mRNA of the gap junction protein Cx43 promote tissue repair in a variety of different wounds. Delivery of the antisense drug has most often been achieved by a thermoreversible hydrogel, Pluronic F-127, which is very effective in the short term but does not allow for sustained delivery over several days. For chronic wounds that take a long time to heal, repeated dosing with the drug may be desirable but is not always compatible with conventional treatments such as the weekly changing of compression bandages on venous leg ulcers. Here the coating of collagen scaffolds with antisense oligonucleotides is investigated and a way to provide protection of the oligodeoxynucleotide drug is found in conjunction with sustained release over a 7 d period. This approach significantly reduces the normal foreign body reaction to the scaffold, which induces an increase of Cx43 protein and an inhibition of healing. As a result of the antisense integration into the scaffold, inflammation is reduced with the rate of wound healing and contracture is significantly improved. This coated scaffold approach may be very useful for treating venous leg ulcers and also for providing a sustained release of any other types of oligonucleotide drugs that are being developed.
Collapse
Affiliation(s)
- Daniel J. Gilmartin
- Department of Cell and Developmental Biology University College London London WC1E 6BT UK
| | - Allyson Soon
- Lee Kong Chian School of Medicine Nanyang Technological University 11, Mandalay Road Singapore 308232
| | | | - Anthony R. J. Phillips
- School of Biological Sciences Department of Surgery University of Auckland New Zealand 1010
| | - Suwan N. Jayasinghe
- BioPhysics Group, Institute of Biomedical Engineering Centre for Stem Cells and Regenerative Medicine, and Department of Mechanical Engineering University College London London WC1E 7JE UK
| | - David L. Becker
- Lee Kong Chian School of Medicine Nanyang Technological University 11, Mandalay Road Singapore 308232
- Institute of Medical Biology A*STAR 8A‐ Biomedical grove, Biopolis Singapore 138648
| |
Collapse
|
21
|
Aghazadeh Y, Papadopoulos V. The role of the 14-3-3 protein family in health, disease, and drug development. Drug Discov Today 2015; 21:278-87. [PMID: 26456530 DOI: 10.1016/j.drudis.2015.09.012] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/21/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022]
Abstract
14-3-3 proteins regulate intracellular signaling pathways, such as signal transduction, protein trafficking, cell cycle, and apoptosis. In addition to the ubiquitous roles of 14-3-3 isoforms, unique tissue-specific functions are also described for each isoform. Owing to their role in regulating cell cycle, protein trafficking, and steroidogenesis, 14-3-3 proteins are prevalent in human diseases, such as cancer, neurodegeneration, and reproductive disorders, and, therefore, serve as valuable drug targets. In this review, we summarize the role of 14-3-3 proteins in normal and disease states, with a focus on 14-3-3γ and ɛ. We also discuss drug compounds targeting 14-3-3 proteins and their potential therapeutic uses.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
22
|
Wachi T, Cornell B, Marshall C, Zhukarev V, Baas PW, Toyo-oka K. Ablation of the 14-3-3gamma Protein Results in Neuronal Migration Delay and Morphological Defects in the Developing Cerebral Cortex. Dev Neurobiol 2015; 76:600-14. [PMID: 26297819 DOI: 10.1002/dneu.22335] [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] [Received: 02/05/2015] [Revised: 07/18/2015] [Accepted: 08/18/2015] [Indexed: 11/10/2022]
Abstract
14-3-3 proteins are ubiquitously-expressed and multifunctional proteins. There are seven isoforms in mammals with a high level of homology, suggesting potential functional redundancy. We previously found that two of seven isoforms, 14-3-3epsilon and 14-3-3zeta, are important for brain development, in particular, radial migration of pyramidal neurons in the developing cerebral cortex. In this work, we analyzed the function of another isoform, the protein 14-3-3gamma, with respect to neuronal migration in the developing cortex. We found that in utero 14-3-3gamma-deficiency resulted in delays in neuronal migration as well as morphological defects. Migrating neurons deficient in 14-3-3gamma displayed a thicker leading process stem, and the basal ends of neurons were not able to reach the boundary between the cortical plate and the marginal zone. Consistent with the results obtained from in utero electroporation, time-lapse live imaging of brain slices revealed that the ablation of the 14-3-3gamma proteins in pyramidal neurons slowed down their migration. In addition, the 14-3-3gamma deficient neurons showed morphological abnormalities, including increased multipolar neurons with a thicker leading processes stem during migration. These results indicate that the 14-3-3gamma proteins play an important role in radial migration by regulating the morphology of migrating neurons in the cerebral cortex. The findings underscore the pathological phenotypes of brain development associated with the disruption of different 14-3-3 proteins and will advance the preclinical data regarding disorders caused by neuronal migration defects.
Collapse
Affiliation(s)
- Tomoka Wachi
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| | - Brett Cornell
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| | - Courtney Marshall
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| | - Vladimir Zhukarev
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| | - Kazuhito Toyo-oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129
| |
Collapse
|
23
|
Bax modulates neuronal survival while p53 is unaltered after Cytochrome C induced oxidative stress in the adult olfactory bulb in vivo. Ann Neurosci 2015; 22:19-25. [PMID: 26124546 PMCID: PMC4410523 DOI: 10.5214/ans.0972.7531.220105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/24/2014] [Accepted: 01/21/2015] [Indexed: 12/24/2022] Open
Abstract
Background The granule and periglomerular cells of the olfactory bulb migrate from the sub-ventricular zone (SVZ) as progenitor cell forming the neuronal stream of the rostral olfactory bulb. These cells are characterized by their ability to divide while expressing adult proteins; a phenomenon attributed to the prolonged cell cycle and the regulatory activities of proteins which modulates apoptosis and proliferation in the developing nervous system. Of interest are the proteins concerned with tumor suppression (p53) and cell cycle exit (Bax) and how they regulate survivability of these neurons in the adult system after an induced oxidative stress. Purpose This study sets to investigate the interplay between p53 and Bax in the adult olfactory bulb (periglomerular and granule cell layer), and how these proteins determine proliferation and neuronal survival after Cytochrome C induced-oxidative stress. Also, we demonstrate the effect of the induced-stress threshold on such regulation in vivo. Methods Adult Wistar rats were segregated into three groups. 10 and 20 mg/Kg BW of potassium cyanide (KCN) was administered to the treatment groups for 15 days while the control received normal saline for the same duration. The olfactory bulb was dissected and processed for general histology and immunohistochemistry of p53/Bax in the periglomerular and granule cell layers. Total (Histology) and immunopositive (p53 and Bax) cell count was done using Image J. Subsequently, we determined the analysis of variance with significance set at *P<0.05. Results We observed an increase in cell count for the 10 mg/KgBW treatment; this was characterized by a significant decrease in Bax expression and no change in p53 expression when this treatment group was compared to the control. However, no change was observed in the total cell count for 20 mg/Kg BW treatment for the same duration of exposure. Interestingly, there was also no significant change in Bax and p53 for this treatment when compared with the control. Conclusion Although p53 plays an important role in development of the olfactory bulb neurons, our findings suggests it has little contribution in neuronal cell viability and proliferation in the adult olfactory bulb. No significant change in p53 was observed irrespective of treatment dose and cell count while Bax expression was reduced at 10 mg/Kg BW treatment and was associated with an increased cell count. We conclude that regulation of survival of neurons in the adult olfactory bulb, following induced-oxidative stress was more dependent of the expression of Bax and the threshold of the induced stress rather than p53 expression.
Collapse
|
24
|
Chen Y, Liu JM, Xiong XX, Qiu XY, Pan F, Liu D, Lan SJ, Jin S, Yu SB, Chen XQ. Piperlongumine selectively kills hepatocellular carcinoma cells and preferentially inhibits their invasion via ROS-ER-MAPKs-CHOP. Oncotarget 2015; 6:6406-21. [PMID: 25788268 PMCID: PMC4467445 DOI: 10.18632/oncotarget.3444] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 01/21/2015] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinomas (HCC) are highly malignant and aggressive tumors lack of effective therapeutic drugs. Piperlongumine (PL), a natural product isolated from longer pepper plants, is recently identified as a potent cytotoxic compound highly selective to cancer cells. Here, we reported that PL specifically suppressed HCC cell migration/invasion via endoplasmic reticulum (ER)-MAPKs-CHOP signaling pathway. PL selectively killed HCC cells but not normal hepatocytes with an IC50 of 10-20 µM while PL at much lower concentrations only suppressed HCC cell migration/invasion. PL selectively elevated reactive oxygen species (ROS) in HCC cells, which activated or up-regulated downstream PERK/Ire 1α/Grp78, p38/JNK/Erk and CHOP subsequently. Administration of antioxidants completely abolished PL's effects on cell death and migration/invasion. However, pharmacological inhibition of ER stress-responses or MAPKs signaling pathways with corresponding specific inhibitors only reversed PL's effect on cell migration/invasion but not on cell death. Consistently, knocking-down of CHOP by RNA interference only reversed PL-suppressed HCC cell migration. Finally, PL significantly suppressed HCC development and activated the ER-MAPKs-CHOP signaling pathway in HCC xenografts in vivo. Taken together, PL selectively killed HCC cells and preferentially inhibited HCC cell migration/invasion via ROS-ER-MAPKs-CHOP axis, suggesting a novel therapeutic strategy for the highly malignant and aggressive HCC clinically.
Collapse
Affiliation(s)
- Yong Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ju Mei Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Xin Xiong
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yao Qiu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Pan
- Department of Urology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Di Liu
- Department of Urology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Jue Lan
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Si Jin
- Department of Pharmacology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Shang Bin Yu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Qian Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Pang Y, Chai CR, Gao K, Jia XH, Kong JG, Chen XQ, Vatcher G, Chen JG, Yu ACH. Ischemia preconditioning protects astrocytes from ischemic injury through 14-3-3γ. J Neurosci Res 2015; 93:1507-18. [PMID: 25711139 DOI: 10.1002/jnr.23574] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 01/11/2015] [Accepted: 01/25/2015] [Indexed: 12/28/2022]
Abstract
Stroke is a leading cause of death and disability, and new strategies are required to reduce neuronal injury and improve prognosis. Ischemia preconditioning (IPC) is an intrinsic phenomenon that protects cells from subsequent ischemic injury and might provide promising mechanisms for clinical treatment. In this study, primary astrocytes exhibited significantly less cell death than control when exposed to different durations of IPC (15, 30, 60, or 120 min). A 15-min duration was the most effective IPC to protect astrocytes from 8-hr-ischemia injury. The protective mechanisms of IPC involve the upregulation of protective proteins, including 14-3-3γ, and attenuation of malondialdehyde (MDA) content and ATP depletion. 14-3-3γ is an antiapoptotic intracellular protein that was significantly upregulated for up to 84 hr after IPC. In addition, IPC promoted activation of the c-Jun N-terminal kinase (JNK), extracellular signal-related kinase (ERK)-1/2, p38, and protein kinase B (Akt) signaling pathways. When JNK was specifically inhibited with SP600125, the upregulation of 14-3-3γ induced by IPC was almost completely abolished; however, there was no effect on ATP or MDA levels. This suggests that, even though both energy preservation and 14-3-3γ up-regulation were turned on by IPC, they were controlled by different pathways. The ERK1/2, p38, and Akt signaling pathways were not involved in the 14-3-3γ upregulation and energy preservation. These results indicate that IPC could protect astrocytes from ischemia injury by inducing 14-3-3γ and by alleviating energy depletion through different pathways, suggesting multiple protection of IPC and providing new insights into potential stroke therapies.
Collapse
Affiliation(s)
- Ying Pang
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Chao Rui Chai
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Kai Gao
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Xi Hua Jia
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Jin Ge Kong
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Xiao Qian Chen
- Department of Pathophysiology, Ministry of Education and Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Greg Vatcher
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Jian Guo Chen
- Key Laboratory of Biomembrane and Membrane Bioengineering, Key Laboratory of Cell Proliferation and Differentiation, Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Albert Cheung Hoi Yu
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.,Laboratory of Translational Medicine, Institute of Systems Biomedicine, Peking University, Beijing, China
| |
Collapse
|
26
|
The protective effect of epoxyeicosatrienoic acids on cerebral ischemia/reperfusion injury is associated with PI3K/Akt pathway and ATP-sensitive potassium channels. Neurochem Res 2014; 40:1-14. [PMID: 25366463 DOI: 10.1007/s11064-014-1456-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/11/2014] [Accepted: 10/09/2014] [Indexed: 12/27/2022]
Abstract
Epoxyeicosatrienoic acids (EETs), the cytochrome P450 epoxygenase metabolite of arachidonic acid, have been demonstrated to have neuroprotective effect. Phosphatidylinositol 3-kinase (PI3K)/Akt and ATP-sensitive potassium (KATP) channels are thought to be important factors that mediate neuroprotection. However, little is known about the role of PI3K/Akt and KATP channels in brain after EETs administration. In vitro experiment, oxygen-glucose deprivation (OGD) was performed in cultured rat cerebral microvascular smooth muscle cells (SMCs) for 4 h. The effect of 14,15-EET on OGD induced cell apoptosis was examined after reoxygenation. Western blot and real-time PCR were used to analyze the expression of Kir6.1, SUR2B (two subunits of KATP channels) and p-Akt on cerebral microvascular SMCs. In vivo experiments, we use 12-(3-adamantan-1-yl-ureido)-dodecanoic acid [AUDA, a specific soluble epoxide hydrolase (sEH) inhibitor] to confirm the effect of EETs indirectly. Rats were injected intraperitoneally with AUDA before being subjected to middle cerebral artery occlusion (MCAO). We detected the apoptosis and the expression of p-Akt, Kir6.1 and SUR2B in ischemic penumbra. The results showed that EETs protect against cerebral ischemia/reperfusion (I/R) injury and upregulated the expression of p-Akt and Kir6.1 in both of ischemic penumbra and OGD induced cerebral microvascular SMCs. The protective effect was inhibited by Wortmannin (a specific PI3K inhibitor) and Glib (a specific KATP inhibitor) respectively in vitro experiment. In conclusion, these results suggested that the protective effect of EETs on cerebral I/R injury is associated with PI3K/Akt pathway and KATP channels. Furthermore, the PI3K pathway may contribute to mediating KATP channels on cerebral microvascular SMCs.
Collapse
|