1
|
Choudhury S, Anne A, Singh M, Chaillet JR, Mohan KN. DNMT1 Y495C mutation interferes with maintenance methylation of imprinting control regions. Int J Biochem Cell Biol 2024; 169:106535. [PMID: 38281697 DOI: 10.1016/j.biocel.2024.106535] [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: 09/19/2023] [Revised: 01/05/2024] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
Hereditary Sensory and Autonomic Neuropathy Type 1E (HSAN1E) is a rare autosomal dominant neurological disorder due to missense mutations in DNA methyltransferase 1 (DNMT1). To investigate the nature of the dominant effect, we compared methylomes of transgenic R1wtDnmt1 and R1Dnmt1Y495C mouse embryonic stem cells (mESCs) overexpressing WT and the mutant mouse proteins respectively, with the R1 (wild-type) cells. In case of R1Dnmt1Y495C, 15 out of the 20 imprinting control regions were hypomethylated with transcript level dysregulation of multiple imprinted genes in ESCs and neurons. Non-imprinted regions, minor satellites, major satellites, LINE1 and IAP repeats were unaffected. These data mirror the specific imprinting defects associated with transient removal of DNMT1 in mESCs, deletion of the maternal-effect DNMT1o variant in preimplantation mouse embryos, and in part, reprogramming to naïve human iPSCs. This is the first DNMT1 mutation demonstrated to specifically affect Imprinting Control Regions (ICRs), and reinforces the differences in maintenance methylation of ICRs over non-imprinted regions. Consistent with nervous system abnormalities in the HSAN1E disorder and involvement of imprinted genes in normal development and neurogenesis, R1Dnmt1Y495C cells showed dysregulated pluripotency and neuron marker genes, and yielded more slender, shorter, and extensively branched neurons. We speculate that R1Dnmt1Y495C cells produce predominantly dimers containing mutant proteins, leading to a gradual and specific loss of ICR methylation during early human development.
Collapse
Affiliation(s)
- Sumana Choudhury
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, BITS Pilani Hyderabad Campus, Hyderabad 500078, India; Centre for Human Disease Research, BITS Pilani Hyderabad Campus, Hyderabad 500078, India
| | - Anuhya Anne
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, BITS Pilani Hyderabad Campus, Hyderabad 500078, India; Centre for Human Disease Research, BITS Pilani Hyderabad Campus, Hyderabad 500078, India
| | - Minali Singh
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, BITS Pilani Hyderabad Campus, Hyderabad 500078, India
| | - John Richard Chaillet
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kommu Naga Mohan
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, BITS Pilani Hyderabad Campus, Hyderabad 500078, India; Centre for Human Disease Research, BITS Pilani Hyderabad Campus, Hyderabad 500078, India.
| |
Collapse
|
2
|
Li T, Su D, Lu H, Gao Y, Liu Y, Wang S, Hou Y, Qin K, Que X, Chen X, Qin B, Wang Z, Deng Y. Recombinant human brain natriuretic peptide attenuates ischemic brain injury in mice by inhibiting oxidative stress and cell apoptosis via activation of PI3K/AKT/Nrf2/HO-1 pathway. Exp Brain Res 2023; 241:2751-2763. [PMID: 37847304 DOI: 10.1007/s00221-023-06716-4] [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: 06/25/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
Ischemic stroke followed by cerebral artery occlusion is a main cause of chronic disability worldwide. Recombinant human brain natriuretic peptide (rhBNP) has been reported to alleviate sepsis-induced cognitive dysfunction and brain I/R injury. However, the function and molecular mechanisms of rhBNP in ischemic brain injury have not been clarified. For establishment of an animal model of ischemic brain injury, C57BL/6 mice were treated with middle cerebral artery occlusion (MCAO) surgery for 1 h and reperfusion for 24 h. After subcutaneous injection of rhBNP into model mice, neurologic deficits were assessed by evaluating behavior of mice according to Longa scoring system, and TTC staining was utilized to determine the brain infarct size of mice. The levels of oxidative stress markers, superoxide dismutase (SOD), catalase (CAT), glutathione (GSH) and malondialdehyde (MDA), were detected in hippocampal tissues of mice by corresponding kits. Cell apoptosis in hippocampus tissues was examined by TUNEL staining. Protein levels of antioxidant enzymes (HO-1 and NQO1) in cerebral cortex, apoptotic markers (Bax, Bcl-2, and cleaved caspase), and PI3K/AKT pathway-associated factors in hippocampus were tested by western blot analysis. The results revealed that injection of rhBNP decreased neurologic deficit scores, the percent of brain water content, and infarct volume. Additionally, rhBNP downregulated MDA level, upregulated the levels of SOD, CAT, and GSH in hippocampus of mice, and increased protein levels of HO-1 and NQO1 in the cortex. Cell apoptosis in hippocampus tissues of model mice was inhibited by rhBNP which was shown as the reduced TUNEL-positive cells, the decreased Bax, cleaved caspase-3, and cleaved caspase-9 protein levels, and the enhanced Bcl-2 protein level. In addition, rhBNP treatment activated the PI3K/AKT signaling pathway and upregulated the protein levels of HO-1 and NRF2. Overall, rhBNP activates the PI3K/AKT/HO-1/NRF2 pathway to attenuate ischemic brain injury in mice after MCAO by suppression of cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Tong Li
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - DaJing Su
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - HuaWen Lu
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YunQing Gao
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YongGang Liu
- Department of Urology Surgery, Nanning Second People's Hospital, Nanning, 530031, China
| | - ShaoHua Wang
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YuTing Hou
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - KeMin Qin
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - XianTing Que
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - XiaoPing Chen
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - BaiLing Qin
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - ZiJun Wang
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - Yan Deng
- Department of Medical Records, Nanning Second People's Hospital, No. 13, Dancun Road, Jiangnan District, Nanning, 530031, Guangxi, China.
| |
Collapse
|
3
|
Tao H, Li L, Dong L, Chen H, Shan X, Zhuge L, Lou H. Growth differentiation factor 7 pretreatment enhances the therapeutic capacity of bone marrow-derived mesenchymal stromal cells against cerebral ischemia-reperfusion injury. Chem Biol Interact 2023; 386:110779. [PMID: 37879595 DOI: 10.1016/j.cbi.2023.110779] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/13/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) transplantation is a promising therapeutic strategy for cerebral ischemia/reperfusion (I/R) injury; however, the clinical outcome is barely satisfactory and demands further improvement. The present study aimed to investigate whether preconditioning of BMSCs by recombinant human growth differentiation factor 7 (rhGDF7) could enhance its therapeutic capacity against cerebral I/R injury. Mouse BMSCs and primary neurons were co-cultured and exposed to oxygen glucose deprivation/reperfusion (OGD/R) stimulation. To investigate the role of exosomal microRNA-369-3p (miR-369-3p), inhibitors, RNAi and the miR-369-3p antagomir were used. Meanwhile, mice were intravenously injected with rhGDF7-preconditioned BMSCs and then received cerebral I/R surgery. Markers of inflammation, oxidative stress and neural damage were evaluated. To inhibit AMP-activated protein kinase (AMPK), compound C was used in vivo and in vitro. Compared with cell-free transwell or vehicle-preconditioned BMSCs, rhGDF7-preconditioned BMSCs significantly prevented OGD/R-induced inflammation, oxidative stress and neural damage in vitro. Meanwhile, rhGDF7-preconditioned BMSCs could prevent I/R-induced cerebral inflammation and oxidative stress in vivo. Mechanistically, rhGDF7 preconditioning significantly increased exosomal miR-369-3p expression in BMSCs and then transferred exosomal miR-369-3p to primary neurons, where it bound to phosphodiesterase 4 D (Pde4d) 3'-UTR and downregulated PDE4D expression, thereby preventing I/R-induced inflammation, oxidative stress and neural damage through activating AMPK pathway. Our study identify GDF7 pretreatment as a promising adjuvant reagent to improve the therapeutic potency of BMSCs for cerebral I/R injury and ischemic stroke.
Collapse
Affiliation(s)
- Hongmiao Tao
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Lin Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Lihua Dong
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Haohao Chen
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Xiaoyun Shan
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Jinhua, 321000, Zhejiang, China
| | - Lujie Zhuge
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Hongqiang Lou
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China.
| |
Collapse
|
4
|
Li Y, Gao J, Liu C, Bu N, Zhan S, Wu H, Zhang R, Sun H, Fan H. USP22 knockdown protects against cerebral ischemia/reperfusion injury via destabilizing PTEN protein and activating the mTOR/TFEB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3163-3175. [PMID: 37191727 DOI: 10.1007/s00210-023-02524-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Ubiquitin-specific protease 22 (USP22) expression was reported to be increased in response to ischemic brain damage, but the biological role and underlying mechanism remain little understood. USP22 shRNA was intravenously injected into the mouse brain, and then a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was constructed, and the infarct volume, neurobehavioral deficit score, cell apoptosis, oxidative stress, and autophagy in vivo were evaluated. Oxygen-glucose deprivation/reperfusion (OGD/R) treated pheochromocytoma-12 (PC12) cells were used as an in vitro model of ischemia/reperfusion. The effects of USP22 on proliferation, apoptosis, oxidative stress, and autophagy were explored by CCK-8, flow cytometry, ELISA, and Western blot assays. The relationship between USP22 and the phosphatase and tensin homolog (PTEN) was measured by Co-IP and Western blot assays. Both USP22 and PTEN were highly expressed in MCAO/R mouse brain tissues and OGD/R-induced PC12 cells. In vitro, USP22 knockdown strongly improved OGD/R-mediated changes in cell viability, apoptosis, oxidative stress, and lactate dehydrogenase (LDH) production in PC12 cells. USP22 bound to PTEN and stabilized PTEN expression by decreasing its ubiquitination. PTEN overexpression reversed the promoting effect of USP22 knockdown on cell viability and the inhibitory effects of USP22 knockdown on apoptosis, oxidative stress, and LDH release rate in PC12 cells subjected to OGD/R. PTEN silencing elevated the protein levels of p62, p-mTOR, TFEB, and LAMP1 and reduced the protein levels of LC3-II/LC3-I. USP22 expression levels were negatively correlated with mTOR expression levels, and USP22-shRNA-mediated expression of p62, p-mTOR, TFEB, and LAMP1 was reversed by rapamycin, an inhibitor of mTOR. In vivo, USP22 silencing significantly alleviated infarct volume, neurobehavioral impairments, cell apoptosis, oxidative stress, and autophagy in MCAO/R mice. USP22 knockdown exerts neuroprotective effects in cerebral ischemia/reperfusion injury by downregulating PTEN and activating the mTOR/TFEB pathway.
Collapse
Affiliation(s)
- Yanling Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China.
| | - Jing Gao
- Department of Anesthesiology, The First Hospital of YuLin, YuLin, 719000, Shaanxi Province, China
| | - Chuntian Liu
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an , Shaanxi Province, China
| | - Ning Bu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| | - Shuqin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| | - Haiqin Wu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| | - Ru Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| | - Hong Sun
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| | - Hong Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, Shaanxi Province, China
| |
Collapse
|
5
|
Zhang H, Du D, Gao X, Tian X, Xu Y, Wang B, Yang S, Liu P, Li Z. PFT-α protects the blood-brain barrier through the Wnt/β-catenin pathway after acute ischemic stroke. Funct Integr Genomics 2023; 23:314. [PMID: 37777676 DOI: 10.1007/s10142-023-01237-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
The dysfunction of blood-brain barrier (BBB) plays a pivotal role in brain injury and subsequent neurological deficits of ischemic stroke. The current study aimed to examine the potential correlation between p53 inhibition and the neuroprotective effect of on the BBB. Rat middle cerebral artery occlusion and reperfusion model (MCAO/R) and oxygen-glucose deprivation/re-oxygenation model (OGD/R) were employed to simulate cerebral ischemia-reperfusion (CI/R) injury occurrence in vivo and in vitro. mNSS and TTC staining were applied to evaluate neurological deficits and brain infarct volumes. Evans blue (EB) staining was carried out to examine the permeability of BBB. RT-qPCR and Western blot to examine the mRNA and protein levels. Cell viabilities were detected by CCK-8. Flow cytometry and ELISA assay were employed to examine apoptosis and neuroinflammation levels. TEER value and sodium fluorescein were carried out to explore the permeability of HBMEC cells. PFT-α inhibited P53 and promoted the expression of β-catenin and cyclin D1, which were reversed by DKK1. PFT-α inhibited neurological deficits, brain infarct volume, neuroinflammation, apoptosis, and BBB integrity than the MCAO/R rats; however, this inhibition was reversed by DKK1. PFT-α promoted OGD/R-induced cell viability in NSCs, and suppressed inflammation and apoptosis, but DKK1 weakened the effect of PFT-α. PFT-α increased OGD/R-induced TEER values in cerebrovascular endothelial cells, inhibited sodium fluorescein permeability, and increased the mRNA levels of tight junction protein, but they were all attenuated by DKK1. PFT-α protects the BBB after acute ischemic stroke via the Wnt/β-catenin pathway, which in turn improves neurological function.
Collapse
Affiliation(s)
- Haitao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Deyong Du
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Xiaoning Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Xiaoling Tian
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Yongqiang Xu
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Bo Wang
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Shoujuan Yang
- Department of Cardiology, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| | - Pengfei Liu
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| | - Zefu Li
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| |
Collapse
|
6
|
Tao H, Dong L, Shan X, Li L, Chen H. MicroRNA-32-3p facilitates cerebral ischemia/reperfusion injury through inhibiting Cab39/AMPK. Int Immunopharmacol 2023; 121:110504. [PMID: 37379707 DOI: 10.1016/j.intimp.2023.110504] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023]
Abstract
Oxidative stress is a key pathogenic factor of cerebral ischemia/reperfusion (I/R) injury. MicroRNA-32-3p (miR-32-3p) plays critical roles in regulating ischemic diseases; however, its role in oxidative stress and cerebral I/R injury remains elusive. Primary cortical neurons and rats were treated with the agomir, antagomir and matched controls of miR-32-3p, and then received oxygen glucose deprivation/reperfusion (OGD/R) or I/R stimulation. To investigate the involvement of AMP-activated protein kinase (AMPK) and calcium-binding protein 39 (Cab39), a pharmacological inhibitor and small interfering RNA were used in vivo and in vitro. Herein, we found that miR-32-3p was upregulated in OGD/R-treated neurons and I/R-injured brains, and that inhibiting miR-32-3p by the miR-32-3p antagomir dramatically alleviated oxidative stress and neural death in OGD/R-stimulated primary cortical neurons. Conversely, overexpressing miR-32-3p by the miR-32-3p agomir further aggravated OGD/R-induced neural death and oxidative damage in primary cortical neurons. Meanwhile, we observed that the miR-32-3p antagomir prevented, while the miR-32-3p agomir facilitated neural death, oxidative damage and cerebral I/R injury in vivo. Mechanistically, miR-32-3p bound to the 3'-untranslated regions of Cab39, inhibited its protein level and subsequently inactivated AMPK. Conversely, treatment with the miR-32-3p antagomir upregulated Cab39 and activated AMPK, thereby attenuating oxidative damage and cerebral I/R injury. Moreover, inhibiting AMPK or Cab39 dramatically blocked the miR-32-3p antagomir-mediated beneficial effects against cerebral I/R injury in vivo and in vitro. miR-32-3p plays critical roles in neural death and oxidative damage upon I/R stimulation, and it is a novel target to treat cerebral I/R injury.
Collapse
Affiliation(s)
- Hongmiao Tao
- Medical College, Jinhua Polytechnic, Jinhua 321017, Zhejiang, China.
| | - Lihua Dong
- Medical College, Jinhua Polytechnic, Jinhua 321017, Zhejiang, China
| | - Xiaoyun Shan
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang, China
| | - Lin Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Haohao Chen
- Medical College, Jinhua Polytechnic, Jinhua 321017, Zhejiang, China
| |
Collapse
|