1
|
Bailly C, Degand C, Laine W, Sauzeau V, Kluza J. Implication of Rac1 GTPase in molecular and cellular mitochondrial functions. Life Sci 2024; 342:122510. [PMID: 38387701 DOI: 10.1016/j.lfs.2024.122510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Rac1 is a member of the Rho GTPase family which plays major roles in cell mobility, polarity and migration, as a fundamental regulator of actin cytoskeleton. Signal transduction by Rac1 occurs through interaction with multiple effector proteins, and its activity is regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). The small protein is mainly anchored to the inner side of the plasma membrane but it can be found in endocellular compartments, notably endosomes and cell nuclei. The protein localizes also into mitochondria where it contributes to the regulation of mitochondrial dynamics, including both mitobiogenesis and mitophagy, in addition to signaling processes via different protein partners, such as the proapoptotic protein Bcl-2 and chaperone sigma-1 receptor (σ-1R). The mitochondrial form of Rac1 (mtRac1) has been understudied thus far, but it is as essential as the nuclear or plasma membrane forms, via its implication in regulation of oxidative stress and DNA damages. Rac1 is subject to diverse post-translational modifications, notably to a geranylgeranylation which contributes importantly to its mitochondrial import and its anchorage to mitochondrial membranes. In addition, Rac1 contributes to the mitochondrial translocation of other proteins, such as p53. The mitochondrial localization and functions of Rac1 are discussed here, notably in the context of human diseases such as cancers. Inhibitors of Rac1 have been identified (NSC-23766, EHT-1864) and some are being developed for the treatment of cancer (MBQ-167) or central nervous system diseases (JK-50561). Their effects on mtRac1 warrant further investigations. An overview of mtRac1 is provided here.
Collapse
Affiliation(s)
- Christian Bailly
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; OncoWitan, Consulting Scientific Office, Lille (Wasquehal) 59290, France.
| | - Claire Degand
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - William Laine
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Vincent Sauzeau
- Université de Nantes, CHU Nantes, CNRS, INSERM, Institut du thorax, Nantes, France
| | - Jérôme Kluza
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| |
Collapse
|
2
|
Yang Z, Jiang Y, Xiao Y, Qian L, Jiang Y, Hu Y, Liu X. Di-Huang-Yin-Zi regulates P53/SLC7A11 signaling pathway to improve the mechanism of post-stroke depression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117226. [PMID: 37748635 DOI: 10.1016/j.jep.2023.117226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 09/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Post-stroke depression (PSD) is a condition characterized by a profoundly depressed mood and diminished interest following a stroke. Di-Huang-Yin-Zi (DHYZ), a traditional Chinese herbal preparation, gained widespread use and shown favorable outcomes in PSD treatment. However, the combination mechanisms of this formula for PSD remain unclear. AIM OF STUDY This study aimed to assess the therapeutic effects of DHYZ extract on rats with PSD and further investigate its underlying mechanism. MATERIALS AND METHODS The active components of DHYZ extract were quantified by the high-performance liquid chromatography-ultraviolet (HPLC-UV). Neurofunctional and depressive-like behavioral tests were performed to assess the neuroprotective effects of DHYZ extract after establishing a PSD rat model. Brain tissue damage volume was assessed using TTC staining, and transmission electron microscopy (TEM) was used to observe the ultrastructural changes of neurons in the prefrontal cortex region, while cell apoptosis was evaluated through TUNEL assay in the prefrontal cortex region of the brain. The effect of DHYZ on ferroptosis markers includes Fe2+, malondialdehyde (MDA), reactive oxygen species (ROS), and glutathione (GSH) were determined in the brain tissue. Moreover, the expression of key proteins or mRNA levels of the P53/SLC7A11 signaling pathway were detected using Western blot or PCR, respectively. Additionally, P53-constructed overexpression vectors were injected to increase the level of P53. In this series of experiments, ferroptosis markers and key factors of the P53/SLC7A11 signaling pathway were evaluated. RESULTS DHYZ extract could increase the sucrose preference of SPT, but decrease the duration of immobility of FST and cortical infarct volume of PSD rats. A TEM study revealed that DHYZ extract improved synaptic ultrastructure in the cortical region of PSD rats. Furthermore, DHYZ treatment effectively decreased ROS and MDA levels, inhibiting the expression of ferroptosis-related markers such as Fe2+, SLC7A11, and GPX4. Additionally, DHYZ promoted the ubiquitination of P53, thus inhibiting its degradation. Notably, AAV-mediated overexpression of P53 reversed the effects of DHYZ on neuroprotection and ferroptosis inhibition in PSD rats. CONCLUSIONS Our results demonstrated that DHYZ extract alleviates the symptoms and enhances the functional capability of PSD rats, mainly by suppressing the ferroptosis through the P53/SLC7A11/GPX4 pathway.
Collapse
Affiliation(s)
- Zhou Yang
- Lianyungang Hospital of Traditional Chinese Medicine, 222004, Lianyungang, China
| | - Yongxia Jiang
- Lianyungang Hospital of Traditional Chinese Medicine, 222004, Lianyungang, China
| | - Yang Xiao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Lihui Qian
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Yongqu Jiang
- Lianyungang Hospital of Traditional Chinese Medicine, 222004, Lianyungang, China
| | - Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Shen Chun-ti Nation-Famous Experts Studio for Traditional Chinese Medicine Inheritance, Changzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu, 213003, China.
| | - Xiaoli Liu
- Lianyungang Hospital of Traditional Chinese Medicine, 222004, Lianyungang, China.
| |
Collapse
|
3
|
Liu A, Yu L, Li X, Zhang K, Zhang W, So KF, Tissir F, Qu Y, Zhou L. Celsr2-mediated morphological polarization and functional phenotype of reactive astrocytes in neural repair. Glia 2023. [PMID: 37186402 DOI: 10.1002/glia.24378] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023]
Abstract
Neural repair is highly influenced by reactive astrocytes. Atypical cadherin Celsr2 regulates neuron development and axon regeneration, while its role in glial cells remains unexplored. In this study, we show that Celsr2 is highly expressed in spinal astrocytes of adult mice, and knockout of Celsr2 results in reactive astrocytes with longer protrusions preferentially orientated towards lesion borders in culture scratch assay and injured spinal cord, and elevation of total and active Cdc42 and Rac1 protein in western blots. Inactivation of Celsr2 enhances calcium influx in reactive astrocytes in time-lapse imaging. Morphological phenotypes of cultured Celsr2-/- astrocytes are rescued by Cdc42 or Rac1 inhibitors. Following spinal cord injury (SCI), Celsr2-/- mice exhibit smaller lesion cavity and glial scar, enhanced fiber regeneration, weaker microglial response, and improved functional recovery than control animals. Similar phenotypes are found in mice with conditional knockout of Celsr2 in astrocytes. In Celsr2-/- mice, astrocyte phenotype is changed and neuroinflammation is alleviated after injury. Inhibiting Cdc42/Rac1 activities compromises astrocyte polarization and the improvement of neural repair and functional recovery in Celsr2-/- mice with SCI. In conclusion, Celsr2 regulates morphological polarization and functional phenotype of reactive astrocytes and inactivating Celsr2 is a potential therapeutic strategy for neural repair.
Collapse
Affiliation(s)
- Aimei Liu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
| | - Lingtai Yu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
| | - Xuejun Li
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
| | - Kejiao Zhang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
| | - Wei Zhang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, People's Republic of China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Fadel Tissir
- Institute of Neuroscience, Developmental Neurobiology, Université catholique de Louvain, Brussels, Belgium
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Yibo Qu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, People's Republic of China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, People's Republic of China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, People's Republic of China
| |
Collapse
|
4
|
Lai AKW, Ng TC, Hung VKL, Tam KC, Cheung CW, Chung SK, Lo ACY. Exacerbated VEGF up-regulation accompanies diabetes-aggravated hemorrhage in mice after experimental cerebral ischemia and delayed reperfusion. Neural Regen Res 2021; 17:1566-1575. [PMID: 34916442 PMCID: PMC8771109 DOI: 10.4103/1673-5374.330612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Reperfusion therapy is the preferred treatment for ischemic stroke, but is hindered by its short treatment window, especially in patients with diabetes whose reperfusion after prolonged ischemia is often accompanied by exacerbated hemorrhage. The mechanisms underlying exacerbated hemorrhage are not fully understood. This study aimed to identify this mechanism by inducing prolonged 2-hour transient intraluminal middle cerebral artery occlusion in diabetic Ins2Akita/+ mice to mimic patients with diabetes undergoing delayed mechanical thrombectomy. The results showed that at as early as 2 hours after reperfusion, Ins2Akita/+ mice exhibited rapid development of neurological deficits, increased infarct and hemorrhagic transformation, together with exacerbated down-regulation of tight-junction protein ZO-1 and up-regulation of blood-brain barrier-disrupting matrix metallopeptidase 2 and matrix metallopeptidase 9 when compared with normoglycemic Ins2+/+ mice. This indicated that diabetes led to the rapid compromise of vessel integrity immediately after reperfusion, and consequently earlier death and further aggravation of hemorrhagic transformation 22 hours after reperfusion. This observation was associated with earlier and stronger up-regulation of pro-angiogenic vascular endothelial growth factor (VEGF) and its downstream phospho-Erk1/2 at 2 hours after reperfusion, which was suggestive of premature angiogenesis induced by early VEGF up-regulation, resulting in rapid vessel disintegration in diabetic stroke. Endoplasmic reticulum stress-related pro-apoptotic C/EBP homologous protein was overexpressed in challenged Ins2Akita/+ mice, which suggests that the exacerbated VEGF up-regulation may be caused by overwhelming endoplasmic reticulum stress under diabetic conditions. In conclusion, the results mimicked complications in patients with diabetes undergoing delayed mechanical thrombectomy, and diabetes-induced accelerated VEGF up-regulation is likely to underlie exacerbated hemorrhagic transformation. Thus, suppression of the VEGF pathway could be a potential approach to allow reperfusion therapy in patients with diabetic stroke beyond the current treatment window. Experiments were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong [CULATR 3834-15 (approval date January 5, 2016); 3977-16 (approval date April 13, 2016); and 4666-18 (approval date March 29, 2018)].
Collapse
Affiliation(s)
- Angela Ka Wai Lai
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Tsz Chung Ng
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Victor Ka Lok Hung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Ka Cheung Tam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Chi Wai Cheung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Sookja Kim Chung
- Macau University of Science and Technology, Taipa, Macau Special Administration Region; School of Biomedical Sciences, The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| |
Collapse
|
5
|
Ping F, Zhang C, Wang X, Wang Y, Zhou D, Hu J, Chen Y, Ling J, Zhou J. Cx32 inhibits the autophagic effect of Nur77 in SH-SY5Y cells and rat brain with ischemic stroke. Aging (Albany NY) 2021; 13:22188-22207. [PMID: 34551394 PMCID: PMC8507301 DOI: 10.18632/aging.203526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
The pathogenesis of cerebral ischemia-reperfusion (I/R) is complex. Cx32 expression has been reported to be up-regulated in ischemic lesions of aged human brain. Nevertheless, the function of Cx32 during cerebral I/R is poorly understood. Autophagy is of vital importance in the pathogenesis of cerebral I/R. In the current study, we found that oxygen-glucose deprivation/reoxygenation (OGD/R) or I/R insult significantly induced the up-regulation of Cx32 and activation of autophagy. Inhibition of Cx32 alleviated OGD/R or I/R injury, and further activated autophagy. In addition, Nur77 expression was found to be up-regulated after OGD/R or I/R. After inhibiting Cx32, the expression of Nur77 was further increased and Nur77 was translocated from nucleus to mitochondrial. Inhibition of Cx32 also activated mitophagy by promoting autophagosome formation and up-regulating the expression of mitochondrial autophagy marker molecules. Of note, in the siNur77-transfected cells, the number of dysfunctional mitochondrial was increased, and mitophagy was suppressed, which aggravated OGD/R-induced neuronal injury. In conclusion, Cx32 might act as a regulatory factor of Nur77 controlling neuronal autophagy in the brains. Understanding the mechanism of this regulatory pathway will provide new insight into the role Cx32 and Nur77 in cerebral ischemia, offering new opportunities for therapeutics.
Collapse
Affiliation(s)
- Fengfeng Ping
- Department of Reproductive Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Chao Zhang
- Department of Reproductive Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Xue Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yan Wang
- Department of Good Clinical Practice, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Danli Zhou
- Department of Good Clinical Practice, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jing Hu
- Department of Good Clinical Practice, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Yanhua Chen
- Department of Good Clinical Practice, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jingjing Ling
- Department of Good Clinical Practice, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jia Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
6
|
Wang Z, Xia P, Hu J, Huang Y, Zhang F, Li L, Wang E, Guo Q, Ye Z. LncRNA MEG3 Alleviates Diabetic Cognitive Impairments by Reducing Mitochondrial-Derived Apoptosis through Promotion of FUNDC1-Related Mitophagy via Rac1-ROS Axis. ACS Chem Neurosci 2021; 12:2280-2307. [PMID: 33843209 DOI: 10.1021/acschemneuro.0c00682] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction and elevated ROS generation are predominant contributors of neuronal death that is responsible for the diabetes-related cognitive impairments. Emerging evidence has demonstrated that long noncoding RNA-MEG3 can serve as an important regulator in the pathogenesis of diabetes. However, the underlying mechanisms remain to be further clarified. Here, it was observed that MEG3 was significantly down-regulated in STZ (streptozotocin)-induced diabetic rats. MEG3 overexpression noticeably improved diabetes-induced cognitive dysfunctions, accompanied by the abatement of Rac1 activation and ROS production, as well as the inhibition of mitochondria-associated apoptosis. Furthermore, either MEG3 overexpression or Rac1 inhibition promoted FUNDC1 dephosphorylation and suppressed oxidative stress and neuro-inflammation. Similarly, in vitro studies confirmed that hyperglycemia also down-regulated MEG3 expression in PC12 cells. MEG3 reintroduction protected PC12 cells against hyperglycemia-triggered neurotoxicity by improving mitochondrial fitness and repressing mitochondria-mediated apoptosis. Moreover, these neuroprotective effects of MEG3 relied on FUNDC1-related mitophagy, since silencing of FUNDC1 abolished these beneficial outcomes. Additionally, MEG3 rescued HG-induced neurotoxicity was involved in inhibiting Rac1 expression via interaction with Rac1 3'UTR. Conversely, knockdown of MEG3 showed opposite effects. NSC23766, a specific inhibitor of Rac1, fully abolished harmful effects of MEG3 depletion. Consistently, knockdown of Rac1 potentiated FUNDC1-associated mitophagy. Meanwhile, colocalization of Rac1 and FUNDC1 was found in mitochondria under hyperglycemia, which was interrupted by MEG3 overexpression. Furthermore, silencing of Rac1 promoted PGAM5 expression, and FUNDC1 strongly interacted with LC3 in Rac1-deleted cells. Altogether, our findings suggested that the Rac1/ROS axis may be a downstream signaling pathway for MEG3-induced neuroprotection, which was involved in FUNDC1-associated mitophagy.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou 570311, China
| | - Pingping Xia
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Jie Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
| | - Fan Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| |
Collapse
|
7
|
Liu Y, Hu XB, Zhang LZ, Wang Z, Fu R. Knockdown of Arginyl-tRNA Synthetase Attenuates Ischemia-Induced Cerebral Cortex Injury in Rats After Middle Cerebral Artery Occlusion. Transl Stroke Res 2021; 12:147-163. [PMID: 32221863 PMCID: PMC7803708 DOI: 10.1007/s12975-020-00809-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
Some researchers have previously shown that RNAi knockdown of arginyl-tRNA synthetase (ArgRS) before or after a hypoxic injury can rescue animals from death, based on the model organism, C. elegans. However, there has been no study on the application of arginyl-tRNA synthetase knockdown in treating mammalian ischemic stroke, and its potential mechanism and effect on ischemic brain damage are still unknown. Here, we focused on the Rars gene, which encodes an arginyl-tRNA synthetase, and examined the effects of Rars knockdown in a permanent middle cerebral artery occlusion model in rats. To achieve this aim, adult male Sprague-Dawley (SD) rats were given right cerebral cortex injections of short hairpin RNA (shRNA) adenovirus (AV) particles to knock down arginyl-tRNA synthetase, and a non-targeting control (NTC) vector or phosphate-buffered solution served as the controls. After 4 days, the rats were exposed to permanent middle cerebral artery occlusion (pMCAO). Then, the right cerebral cortex level of arginyl-tRNA synthetase was examined, and the effects of the Rars knockdown were evaluated by differences in infarction volume, oxidative stress, blood-brain barrier, mitochondrial function, and glucose metabolism at 1 day and 3 days after MCAO. The injection of shRNA adenovirus particles successfully suppressed the expression of arginyl-tRNA synthetase in the cerebral cortex. We observed an improvement in oxidative stress, mitochondrial function, and glucose utilization and a reduction in brain edema compared with the non-targeting control rats with suppressed expression of arginyl-tRNA synthetase mRNA in the ipsilateral ischemic cortex of the brain. Our findings indicate that knockdown of arginyl-tRNA synthetase in the cerebral cortex exerted neuroprotective effects, which were achieved not only by the improvement of oxidative stress and glucose utilization but also by the maintenance of mitochondrial morphological integrity and the preservation of mitochondrial function. Knockdown of ArgRS administration could be a promising approach to protect ischemic stroke.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xue-Bin Hu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Li-Zhi Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zi Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
8
|
Kim MH, Kwon B, Kim KS, Kim MS, Kim MJ, Kim HJ, Choi DI, Park M, Kim M, Shin MK, Jeong SI, Yu KY, Kim J. Galuteolin, identified in the extract of thymus quinquecostatus flowers, is involved in inhibiting melanin biosynthesis in B16/F10 melanoma cells. Nat Prod Res 2020; 35:5389-5391. [PMID: 32419489 DOI: 10.1080/14786419.2020.1768091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
To enhance the skin whitening effect, tyrosinase activity and melanin biosynthesis needs to be suppressed in the skin. To achieve this goal, we examined the extract of Thymus quinquecostatus flowers, and identified a functional ingredient, galuteolin. Galuteolin effectively inhibited melanin biosynthesis in B16/F10 cells, partially suppressing tyrosinase activity. Therefore, this study suggests that galuteolin can be used as a cosmetic ingredient for skin whitening.
Collapse
Affiliation(s)
- Mi-Hee Kim
- Jeonju AgroBio-Materials Institute, Jeonju-si, Korea
| | - Bora Kwon
- Jeonju AgroBio-Materials Institute, Jeonju-si, Korea
| | | | | | | | | | | | | | | | | | | | - Kang-Yeol Yu
- Jeonju AgroBio-Materials Institute, Jeonju-si, Korea
| | - Jiyoung Kim
- Jeonju AgroBio-Materials Institute, Jeonju-si, Korea
| |
Collapse
|
9
|
Abstract
Glucose-induced (physiological) insulin secretion from the islet β-cell involves interplay between cationic (i.e., changes in intracellular calcium) and metabolic (i.e., generation of hydrophobic and hydrophilic second messengers) events. A large body of evidence affirms support for novel regulation, by G proteins, of specific intracellular signaling events, including actin cytoskeletal remodeling, transport of insulin-containing granules to the plasma membrane for fusion, and secretion of insulin into the circulation. This article highlights the following aspects of GPCR-G protein biology of the islet. First, it overviews our current understanding of the identity of a wide variety of G protein regulators and their modulatory roles in GPCR-G protein-effector coupling, which is requisite for optimal β-cell function under physiological conditions. Second, it describes evidence in support of novel, noncanonical, GPCR-independent mechanisms of activation of G proteins in the islet. Third, it highlights the evidence indicating that abnormalities in G protein function lead to islet β-cell dysregulation and demise under the duress of metabolic stress and diabetes. Fourth, it summarizes observations of potential beneficial effects of GPCR agonists in preventing/halting metabolic defects in the islet β-cell under various pathological conditions (e.g., metabolic stress and inflammation). Lastly, it identifies knowledge gaps and potential avenues for future research in this evolving field of translational islet biology. Published 2020. Compr Physiol 10:453-490, 2020.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Center for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
10
|
Xia P, Zhang F, Yuan Y, Chen C, Huang Y, Li L, Wang E, Guo Q, Ye Z. ALDH 2 conferred neuroprotection on cerebral ischemic injury by alleviating mitochondria-related apoptosis through JNK/caspase-3 signing pathway. Int J Biol Sci 2020; 16:1303-1323. [PMID: 32210721 PMCID: PMC7085232 DOI: 10.7150/ijbs.38962] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Past studies have indicated that the dysregulation of Aldehyde dehydrogenase 2 (ALDH2) is related to the pathogenesis of acute stroke. However, the underlying mechanisms of ALDH2-mediated acute stroke are still not well understood. Thus, our study was designed to explore the influence of ALDH2 in acute stroke and determine whether its related mechanisms are involved in regulating mitochondria-associated apoptosis modulating JNK/caspase-3 pathway. In vitro analysis on the gain and loss of ALDH2 and JNK function were performed to explore its influence on OGD/R injury and relevant signaling pathways. Our findings suggested that ALDH2 expression was significantly down-regulated in rats suffering from acute stroke and also in primary cortical cultured neurons and PC12 cells upon OGD/R stimulation. ALDH2 overexpression markedly decreased infarct size and improved neurological outcomes. Furthermore, ALDH2 overexpression significantly suppressed stroke-induced mitochondria-associated apoptosis and inhibited p-JNK activation and p-JNK/caspase-3 complex formation. Similarly, in in vitro OGD/R models, ALDH2 reintroduction not only promoted cellular viability and moderated LDH release, but also inhibited mitochondria-related apoptosis. Moreover, JNK inhibition relieved OGD/R-induced cellular injury and apoptosis while JNK activation aggravated them. Furthermore, ALDH2 overexpression and JNK inhibition significantly reduced caspase-3 activation and transcription which was triggered by OGD/R damage. Caspase-3 activation and transcription also re-elevated during activation of JNK in ALDH2-reintroduced cells. Finally, ChIP assay revealed that p-JNK was bound to caspase-3 promoter. Collectively, ALDH2 overexpression led to a significant reduction in mitochondria-related apoptosis via JNK-mediated caspase-3 activation and transcription in both in vitro and in vivo cerebral ischemia models.
Collapse
Affiliation(s)
- Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Fan Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Yajing Yuan
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Cheng Chen
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| |
Collapse
|
11
|
Xia PP, Zhang F, Chen C, Wang ZH, Wang N, Li LY, Guo QL, Ye Z. Rac1 relieves neuronal injury induced by oxygenglucose deprivation and re-oxygenation via regulation of mitochondrial biogenesis and function. Neural Regen Res 2020; 15:1937-1946. [PMID: 32246643 PMCID: PMC7513980 DOI: 10.4103/1673-5374.280325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Certain microRNAs (miRNAs) can function as neuroprotective factors after reperfusion/ischemia brain injury. miRNA-142-3p can participate in the occurrence and development of tumors and myocardial ischemic injury by negatively regulating the activity of Rac1, but it remains unclear whether miRNA-142-3p also participates in cerebral ischemia/reperfusion injury. In this study, a model of oxygen-glucose deprivation/re-oxygenation in primary cortical neurons was established and the neurons were transfected with miR-142-3p agomirs or miR-142-3p antagomirs. miR-142-3p expression was down-regulated in neurons when exposed to oxygen-glucose deprivation/re-oxygenation. Over-expression of miR-142-3p using its agomir remarkably promoted cell death and apoptosis induced by oxygen-glucose deprivation/re-oxygenation and improved mitochondrial biogenesis and function, including the expression of peroxisome proliferator-activated receptor-γ coactivator-1α, mitochondrial transcription factor A, and nuclear respiratory factor 1. However, the opposite effects were produced if miR-142-3p was inhibited. Luciferase reporter assays verified that Rac Family Small GTPase 1 (Rac1) was a target gene of miR-142-3p. Over-expressed miR-142-3p inhibited NOX2 activity and expression of Rac1 and Rac1-GTPase (its activated form). miR-142-3p antagomirs had opposite effects after oxygen-glucose deprivation/re-oxygenation. Our results indicate that miR-142-3p down-regulates the expression and activation of Rac1, regulates mitochondrial biogenesis and function, and inhibits oxygen-glucose deprivation damage, thus exerting a neuroprotective effect. The experiments were approved by the Committee of Experimental Animal Use and Care of Central South University, China (approval No. 201703346) on March 7, 2017.
Collapse
Affiliation(s)
- Ping-Ping Xia
- Department of Anesthesiology, Xiangya Hospital; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan Province, China
| | - Fan Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Cheng Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhi-Hua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan Province, China
| | - Na Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Long-Yan Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qu-Lian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
12
|
Cheng X, Zhang F, Li J, Wang G. Galuteolin attenuates cerebral ischemia/reperfusion injury in rats via anti-apoptotic, anti-oxidant, and anti-inflammatory mechanisms. Neuropsychiatr Dis Treat 2019; 15:2671-2680. [PMID: 31571883 PMCID: PMC6754329 DOI: 10.2147/ndt.s215263] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/03/2019] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Galuteolin is a substance extracted and purified from honeysuckle. The purpose of this study was to explore the protective effect of galuteolin on cerebral ischemia-reperfusion injury (CIRI) and reveal its potential molecular mechanism from the perspectives of anti-apoptosis, anti-oxidation, and anti-inflammation. MATERIALS AND METHODS One hundred and fifty rats were randomly divided into five groups: sham group, ischemia-reperfusion (I/R) group, 50 mg/kg galuteolin group, 100 mg/kg galuteolin group, and 200 mg/kg galuteolin group. Middle cerebral artery occlusion (MCAO) was used to establish a rat CIRI model, different doses of galtenolin were intraperitoneal injected 2 hrs after ischemia, and then reperfusion was performed for 24 hrs. Neurological function and cerebral water content were determined, and cerebral infarct volume was evaluated by TTC staining. TUNEL staining was used to detect the apoptosis of nerve cells. Western Blot was used to detect the expressions of Akt, p-Akt, Sod1, Sod2, catalase, caspase-3, Bcl-2, and Bax. Lipid hydrogen peroxide (LPO) was determined by kit assay. The contents of vascular endothelial growth factor (VEGF) and pro-inflammatory cytokines IL-1β and TNF-α were determined by ELISA. RESULTS The results showed that galuteolin could significantly reduce the cerebral infarction volume, neurologic score, and cerebral water content in a dose-dependent manner. In addition, galuteolin obviously reduced the apoptosis rate of nerve cells and the expression levels of caspase-3 and Bax, meanwhile up-regulated the expression levels of p-Akt and Bcl-2. Furthermore, galuteolin apparently inhibited the levels of LPO, Sod1, Sod2, and catalase in the cerebral infarction tissues. Moreover, galuteolin also significantly reduced the levels of pro-inflammatory factors IL-1β and TNF-α in the cerebral infarction tissues. Finally, Galuteolin markedly inhibited the expression of VEGF in cerebral infarction tissues. CONCLUSION Galuteolin exerts neuroprotective effects against CIRI by inhibiting apoptosis, oxidation, and inflammation.
Collapse
Affiliation(s)
- Xue Cheng
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province121001, People’s Republic of China
| | - Fan Zhang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province121001, People’s Republic of China
| | - Jingwei Li
- Department of Neurology, Liaoning Health Industry Group Fuxin Mine General Hospital, Jinzhou City, Liaoning Province121001, People’s Republic of China
| | - Gang Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province121001, People’s Republic of China
| |
Collapse
|
13
|
Ras-Related C3 Botulinum Toxin Substrate 1 Promotes Axonal Regeneration after Stroke in Mice. Transl Stroke Res 2018; 9:506-514. [PMID: 29476448 DOI: 10.1007/s12975-018-0611-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Neurite plasticity is a critical aspect of brain functional recovery after stroke. Emerging data suggest that Ras-related C3 botulinum toxin substrate 1 (Rac1) plays a central role in axonal regeneration in the injured brain, specifically by stimulating neuronal intrinsic growth and counteracting the growth inhibitory signaling that leads to growth cone collapse. Therefore, we investigated the functional role of Rac1 in axonal regeneration after stroke.Delayed treatment with a specific Rac1 inhibitor, NSC 23766, worsened functional recovery, which was assessed by the pellet reaching test from day 14 to day 28 after stroke. It additionally reduced axonal density in the peri-infarct zone, assessed 28 days after stroke, with no effect on brain cavity size or on the number of newly formed cells. Accordingly, Rac1 overexpression using lentivirus promoted axonal regeneration and functional recovery after stroke from day 14 to day 28. Rac1 inhibition led to inactivation of pro-regenerative molecules, including mitogen-activated protein kinase kinase (p-MEK)1/2, LIM domain kinase (LIMK)1, and extracellular signal-regulated kinase (p-ERK)1/2 at 14 days after stroke. Inhibition of Rac1 reduced axonal length and number in cultured primary mouse cortical neurons using microfluidic chambers after oxygen-glucose deprivation (OGD) without affecting cell viability. In contrast, inhibition of Rac1 increased levels of glial fibrillary acidic protein, an extrinsic inhibitory signal for axonal growth, after stroke in vivo and in primary astrocytes after OGD.In conclusion, Rac1 signaling enhances axonal regeneration and improve post-stroke functional recovery in experimental models of stroke.
Collapse
|
14
|
Pan Y, Wang N, Xia P, Wang E, Guo Q, Ye Z. Inhibition of Rac1 ameliorates neuronal oxidative stress damage via reducing Bcl-2/Rac1 complex formation in mitochondria through PI3K/Akt/mTOR pathway. Exp Neurol 2017; 300:149-166. [PMID: 29129468 DOI: 10.1016/j.expneurol.2017.10.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 01/28/2023]
Abstract
Although the neuroprotective effects of Rac1 inhibition have been reported in various cerebral ischemic models, the molecular mechanisms of action have not yet been fully elucidated. In this study, we investigated whether the inhibition of Rac1 provided neuroprotection in a diabetic rat model of focal cerebral ischemia and hyperglycemia-exposed PC-12 cells. Intracerebroventricular administration of lentivirus expressing the Rac1 small hairpin RNA (shRNA) and specific Rac1 inhibitor NSC23766 not only decreased the infarct volumes and improved neurologic deficits with a correlated significant activation of mitochondrial DNA specific proteins, such as OGG1 and POLG, but also elevated Bcl-2 S70 phosphorylation in mitochondria. Furthermore, the levels of p-PI3K, p-Akt and p-mTOR increased, while 8-OHdG, ROS production and Bcl-2/Rac1 complex formation in mitochondria reduced in both Rac1-shRNA- and NSC23766-treated rats. Moreover, to confirm our in vivo observations, inhibition of Rac1 activity by NSC23766 suppressed the interactions between Bcl-2 and Rac1 in the mitochondria of PC-12 cells cultured in high glucose conditions and protected PC-12 cells from high glucose-induced neurotoxicity. More importantly, these beneficial effects of Rac1 inhibition were abolished by PI3K inhibitor LY294002. In contrast to NSC23766 treatment, LY294002 had little effect on the decrement of p-PTEN level. Taken together, these findings revealed novel neuroprotective roles of Rac1 inhibition against cerebral ischemic reperfusion injury in vivo and high glucose-induced neurotoxicity in PC-12 cells in vitro, by reducing Bcl-2/Rac1 complex formation in mitochondria through the activation of PI3K/Akt/mTOR survival pathway.
Collapse
Affiliation(s)
- Yundan Pan
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Na Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Pingping Xia
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - E Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Qulian Guo
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China.
| |
Collapse
|
15
|
The Ubiquitin E3 Ligase TRAF6 Exacerbates Ischemic Stroke by Ubiquitinating and Activating Rac1. J Neurosci 2017; 37:12123-12140. [PMID: 29114077 DOI: 10.1523/jneurosci.1751-17.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/19/2017] [Accepted: 10/27/2017] [Indexed: 11/21/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide. Inflammation, oxidative stress, apoptosis, and excitotoxicity contribute to neuronal death during ischemic stroke; however, the mechanisms underlying these complicated pathophysiological processes remain to be fully elucidated. Here, we found that the expression of tumor necrosis factor receptor-associated factor 6 (TRAF6) was markedly increased after cerebral ischemia/reperfusion (I/R) in mice. TRAF6 ablation in male mice decreased the infarct volume and neurological deficit scores and decreased proinflammatory signaling, oxidative stress, and neuronal death after cerebral I/R, whereas transgenic overexpression of TRAF6 in male mice exhibited the opposite effects. Mechanistically, we demonstrated that TRAF6 induced Rac1 activation and consequently promoted I/R injury by directly binding and ubiquitinating Rac1. Either functionally mutating the TRAF6 ubiquitination site on Rac1 or inactivating Rac1 with a specific inhibitor reversed the deleterious effects of TRAF6 overexpression during I/R injury. In conclusion, our study demonstrated that TRAF6 is a key promoter of ischemic signaling cascades and neuronal death after cerebral I/R injury. Therefore, the TRAF6/Rac1 pathway might be a promising target to attenuate cerebral I/R injury.SIGNIFICANCE STATEMENT Stroke is one of the most severe and devastating neurological diseases globally. The complicated pathophysiological processes restrict the translation of potential therapeutic targets into medicine. Further elucidating the molecular mechanisms underlying cerebral ischemia/reperfusion injury may open a new window for pharmacological interventions to promote recovery from stroke. Our study revealed that ischemia-induced tumor necrosis factor receptor-associated factor 6 (TRAF6) upregulation binds and ubiquitinates Rac1 directly, which promotes neuron death through neuroinflammation and neuro-oxidative signals. Therefore, precisely targeting the TRAF6-Rac1 axis may provide a novel therapeutic strategy for stroke recovery.
Collapse
|
16
|
Kowluru A. Role of G-proteins in islet function in health and diabetes. Diabetes Obes Metab 2017; 19 Suppl 1:63-75. [PMID: 28880478 PMCID: PMC5657296 DOI: 10.1111/dom.13011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/03/2017] [Accepted: 05/12/2017] [Indexed: 12/17/2022]
Abstract
Glucose-stimulated insulin secretion (GSIS) involves interplay between metabolic and cationic events. Seminal contributions from multiple laboratories affirm essential roles for small G-proteins (Rac1, Cdc42, Arf6, Rab27A) in GSIS. Activation of these signalling proteins promotes cytoskeletal remodeling, transport and docking of insulin granules on the plasma membrane for exocytotic secretion of insulin. Evidence in rodent and human islets suggests key roles for lipidation (farnesylation and geranylgeranylation) of these G-proteins for their targeting to appropriate cellular compartments for optimal regulation of effectors leading to GSIS. Interestingly, however, inhibition of prenylation appears to cause mislocalization of non-prenylated, but (paradoxically) activated G-proteins, in "inappropriate" compartments leading to activation of stress kinases and onset of mitochondrial defects, loss in GSIS and apoptosis of the islet β-cell. This review highlights our current understanding of roles of G-proteins and their post-translational lipidation (prenylation) signalling networks in islet function in normal health, metabolic stress (glucolipotoxicity and ER stress) and diabetes. Critical knowledge gaps that need to be addressed for the development of therapeutics to halt defects in these signalling steps in β-cells in models of impaired insulin secretion and diabetes are also highlighted and discussed.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- β-Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, and Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan
| |
Collapse
|
17
|
Suppression of Inner Mitochondrial Membrane Peptidase 2-Like (IMMP2L) Gene Exacerbates Hypoxia-Induced Neural Death Under High Glucose Condition. Neurochem Res 2017; 42:1504-1514. [PMID: 28316022 DOI: 10.1007/s11064-017-2207-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 10/19/2022]
Abstract
It is known that diabetes hyperglycemia enhances cerebral ischemia and reperfusion induced damage. We have previously shown that mutation of inner mitochondrial membrane peptidase 2-like (IMMP2L) increases brain damage caused by transient cerebral ischemia. In this study, we attempt to examine the impact of IMMP2L deficiency on an in vitro model that mimics the diabetic hypoxic conditions. Normal IMMP2L wild type and IMMP2L gene deleted HT22 cells were cultured. Hypoxia was induced under high glucose and acidic conditions with 4 h of oxygen deprivation. Cell viability was assessed by CCK-8 assay and cell death was determined using Annexin V/7-AAD assay. Superoxide production was measured using dihydroethidium staining and mitochondrial membrane potential was detected using JC-1 probe. Suppression of IMMP2L reduced the cell viability, increased the ROS production and decreased the mitochondrial membrane potential. In conclusion, our study demonstrated that deficiency of IMMP2L in cells, cultured under hypoxia, high glucose and acidic conditions, exacerbated neuronal death under a condition that mimics in vivo cerebral ischemia in diabetic condition.
Collapse
|
18
|
Wang Z, Ye Z, Huang G, Wang N, Wang E, Guo Q. Sevoflurane Post-conditioning Enhanced Hippocampal Neuron Resistance to Global Cerebral Ischemia Induced by Cardiac Arrest in Rats through PI3K/Akt Survival Pathway. Front Cell Neurosci 2016; 10:271. [PMID: 27965539 PMCID: PMC5127837 DOI: 10.3389/fncel.2016.00271] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
The purpose of this current study was to evaluate whether improvement of mitochondrial dysfunction was involved in the therapeutic effect of sevoflurane post-conditioning in global cerebral ischemia after cardiac arrest (CA) via the PI3K/Akt pathway. In the first experiment, animals were randomly divided into three groups: a sham group, a CA group, a CA+sevoflurane post-conditioning group (CA+SE). Sevoflurane post-conditioning was achieved by administration of 2.5% sevoflurane for 30 min after resuscitation. Sevoflurane post-conditioning has a significant neuroprotective effect by increasing survival rates and reducing neuronal apoptosis. Additionally, the gene and protein expression of PGC-1α, NRF-1, and TFAM, the master regulators of mitochondrial biogenesis, were up-regulated in the CA+SE group, when compared to the CA group. Similarly, in contrast to the CA group, mitochondria-specific antioxidant enzymes, including heat-shock protein 60 (HSP60), peroxiredoxin 3 (Prx3), and thioredoxin 2 (Trx2) were also increased in the CA+SE group. Finally, administration of sevoflurane ameliorated mitochondrial reactive oxygen species (ROS) formation and maintained mitochondrial integrity. In the second experiment, we investigated the relationship between the PI3K/Akt pathway and mitochondrial biogenesis and mitochondria-specific antioxidant enzymes in sevoflurane-induced neuroprotection. The selective PI3K inhibitor wortmannin not only eliminated the beneficial biochemical processes of sevoflurane by reducing the level of mitochondrial biogenesis-related proteins and aggravating mitochondrial integrity, but also reversed the elevation of mitochondria-specific antioxidant enzymes induced by sevoflurane. Therefore, our data suggested that sevoflurane post-conditioning provides neuroprotection via improving mitochondrial biogenesis and integrity, as well as increasing mitochondria-specific antioxidant enzymes by a mechanism involving the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South UniversityChangsha, China; Department of Anesthesiology, Hainan General HospitalHaikou, China
| | - Zhi Ye
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Guoqing Huang
- Emergency Department, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Na Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - E Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| |
Collapse
|
19
|
Gao S, Mo J, Chen L, Wang Y, Mao X, Shi Y, Zhang X, Yu R, Zhou X. Astrocyte GGTI-mediated Rac1 prenylation upregulates NF-κB expression and promotes neuronal apoptosis following hypoxia/ischemia. Neuropharmacology 2016; 103:44-56. [DOI: 10.1016/j.neuropharm.2015.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
|
20
|
Wu J, Zhang Y, Yang P, Enkhjargal B, Manaenko A, Tang J, Pearce WJ, Hartman R, Obenaus A, Chen G, Zhang JH. Recombinant Osteopontin Stabilizes Smooth Muscle Cell Phenotype via Integrin Receptor/Integrin-Linked Kinase/Rac-1 Pathway After Subarachnoid Hemorrhage in Rats. Stroke 2016; 47:1319-27. [PMID: 27006454 DOI: 10.1161/strokeaha.115.011552] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 02/22/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Recombinant osteopontin (rOPN) has been reported to be neuroprotective in stroke animal models. The purpose of this study is to investigate a potential role and mechanism of nasal administration of rOPN on preserving the vascular smooth muscle phenotype in early brain injury after subarachnoid hemorrhage (SAH). METHODS One hundred and ninety-two male adult Sprague-Dawley rats were used. The SAH model was induced by endovascular perforation. Integrin-linked kinase small interfering RNA was intracerebroventricularly injected 48 hours before SAH. The integrin receptor antagonist fibronectin-derived peptide Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP), focal adhesion kinase inhibitor Fib-14, and Rac-1 inhibitor NSC23766 were administered 1 hour before SAH induction. rOPN was administered via the intracerebroventricular and nasal route after SAH. SAH grade, neurological scores, brain water content, brain swelling, hematoxylin and eosin staining, India ink angiography, Western blots, and immunofluorescence were used to study the mechanisms of rOPN on the vascular smooth muscle phenotypic transformation. RESULTS The marker proteins of vascular smooth muscle phenotypic transformation α-smooth muscle actin decreased and embryonic smooth muscle myosin heavy chain (SMemb) increased significantly at 24 and 72 hours in the cerebral arteries after SAH. rOPN prevented the changes of α-smooth muscle actin and SMemb and significantly alleviated neurobehavioral dysfunction, increased the cross-sectional area and the lumen diameter of the cerebral arteries, reduced the brain water content and brain swelling, and improved the wall thickness of cerebral arteries. These effects of rOPN were abolished by GRGDSP, integrin-linked kinase small interfering RNA, and NSC23766. Intranasal application of rOPN at 3 hours after SAH also reduced neurological deficits. CONCLUSIONS rOPN prevented the vascular smooth muscle phenotypic transformation and improved the neurological outcome, which was possibly mediated by the integrin receptor/integrin-linked kinase/Rac-1 pathway.
Collapse
Affiliation(s)
- Jiang Wu
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Yang Zhang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Peng Yang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Budbazar Enkhjargal
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Anatol Manaenko
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Jiping Tang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - William J Pearce
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Richard Hartman
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Andre Obenaus
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Gang Chen
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA.
| | - John H Zhang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA.
| |
Collapse
|
21
|
Wang Y, Kunit T, Ciotkowska A, Rutz B, Schreiber A, Strittmatter F, Waidelich R, Liu C, Stief CG, Gratzke C, Hennenberg M. Inhibition of prostate smooth muscle contraction and prostate stromal cell growth by the inhibitors of Rac, NSC23766 and EHT1864. Br J Pharmacol 2015; 172:2905-17. [PMID: 25631101 PMCID: PMC4439884 DOI: 10.1111/bph.13099] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Medical therapy of lower urinary tract symptoms (LUTS) suggestive of benign prostatic hyperplasia (BPH) targets smooth muscle contraction in the prostate, or prostate growth. However, current therapeutic options are insufficient. Here, we investigated the role of Rac in the control of smooth muscle tone in human prostates and growth of prostate stromal cells. EXPERIMENTAL APPROACH Experiments were performed using human prostate tissues from radical prostatectomy and cultured stromal cells (WPMY-1). Expression of Rac was examined by Western blot and fluorescence staining. Effects of Rac inhibitors (NSC23766 and EHT1864) on contractility were assessed in the organ bath. The effects of Rac inhibitors were assessed by pull-down, cytotoxicity using a cell counting kit, cytoskeletal organization by phalloidin staining and cell growth using an 5-ethynyl-2'-deoxyuridine assay. KEY RESULTS Expression of Rac1-3 was observed in prostate samples from each patient. Immunoreactivity for Rac1-3 was observed in the stroma, where it colocalized with the smooth muscle marker, calponin. NSC23766 and EHT1864 significantly reduced contractions of prostate strips induced by noradrenaline, phenylephrine or electrical field stimulation. NSC23766 and EHT1864 inhibited Rac activity in WPMY-1 cells. Survival of WPMY-1 cells ranged between 64 and 81% after incubation with NSC23766 (50 or 100 μM) or EHT1864 (25 μM) for 24 h. NSC23766 and EHT1864 induced cytoskeletal disorganization in WPMY-1 cells. Both inhibitors impaired the growth of WPMY-1 cells. CONCLUSIONS AND IMPLICATIONS Rac may be a link connecting the control of prostate smooth muscle tone with proliferation of smooth muscle cells. Improvements in LUTS suggestive of BPH by Rac inhibitors appears possible.
Collapse
Affiliation(s)
- Y Wang
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
- Department of Urology, Zhujiang Hospital, Southern Medical UniversityGuangzhou, China
| | - T Kunit
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
- University Hospital for Urology and AndrologySalzburg, Austria
| | - A Ciotkowska
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - B Rutz
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - A Schreiber
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - F Strittmatter
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - R Waidelich
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - C Liu
- Department of Urology, Zhujiang Hospital, Southern Medical UniversityGuangzhou, China
| | - C G Stief
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - C Gratzke
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - M Hennenberg
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| |
Collapse
|
22
|
Tejada-Simon MV. Modulation of actin dynamics by Rac1 to target cognitive function. J Neurochem 2015; 133:767-79. [PMID: 25818528 DOI: 10.1111/jnc.13100] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/14/2022]
Abstract
The small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in cells. Formation of extensions at the surface of the cell is required for migration and even for cell invasion and metastases. Because an elevated level and hyperactivation of this protein has been associated with metastasis in cancer, direct regulators of Rac1 are currently envisioned as a potential strategy to treat certain cancers. Less research, however, has been done regarding the role of this small GTP-binding protein in brain development, where it has an important role in dendritic spine morphogenesis through the regulation of actin. Alteration of dendritic development and spinogenesis has been often associated with mental disorders. Rac1 is associated with and required for learning and the formation of memories in the brain. Rac1 appears to be dysregulated in certain neurodevelopmental disorders that present all these three alterations: mental retardation, atypical synaptic plasticity and aberrant spine morphology. Thus, to develop novel therapies for rescuing cognitive impairment, a reasonable approach might be to target this protein, Rac1, which plays a pivotal role in directing signals that regulate actin dynamics, which in turn might have an effect in spine cytoarchitecture and synaptic function. It is possible that novel drugs that regulate Rac1 activation and function could modulate actin cytoskeleton and spine dynamics, representing potential candidates to repair intellectual disability in disorders associated with spine abnormalities. Herein, we present a list of the current Rac1 inhibitors that might fulfill this role together with a summary of the latest findings concerning their function as they relate to neuronal studies. While the small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in different type of cells, it appears to be also required for learning and the formation of memories in the brain. Abnormal regulation of this protein has been associated with cognitive disabilities, atypical synaptic plasticity and abnormal morphology of dendritic spines in certain neurodevelopmental disorders. Thus, modulation of Rac1 activity using novel inhibitors might be a strategy to reestablish cognitive function.
Collapse
Affiliation(s)
- Maria V Tejada-Simon
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA.,Department of Biology, University of Houston, Houston, Texas, USA.,Department of Psychology, University of Houston, Houston, Texas, USA.,Biology of Behavior Institute (BoBI), University of Houston, Houston, Texas, USA
| |
Collapse
|