1
|
Cha J, Zeng P, Zong H, Zhao J, Chen J, Zuo H, Zhang B, Shi C, Li J, Hua Q, Wang Z, Hou Y, Zhang R. Single-cell RNA sequencing of neonatal cortical astrocytes reveals versatile cell clusters during astrocyte-neuron conversion. Mol Biol Rep 2025; 52:189. [PMID: 39899158 DOI: 10.1007/s11033-025-10309-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Astrocytes are extensively utilized as starting cells for neuronal conversion. Our previous study discovered that a portion of primary cultured mouse neonatal cortical astrocytes can be directly converted into neurons after exposure to a neurogenic induction condition. Recent in vivo studies have demonstrated astrocyte heterogeneity in terms of their developmental origin, molecular profile, physiology, and functional outputs. We hypothesized that the heterogeneity of primary astrocytes in our study could influence their conversion potential. METHODS AND RESULTS We performed single-cell RNA sequencing on cells harvested at key time points during in vitro astrocyte-to-neuron conversion, specifically on Day 1 and Day 9. Through single-cell RNA sequencing analysis, we identified several subpopulations of astrocytes, labeled as Astrocyte 1 to Astrocyte 3, based on distinct gene expression patterns. Pseudotime trajectory analysis predicted the existence of three distinct cell states throughout the conversion process. Astrocyte 3 exhibited a higher propensity for neuronal conversion, with proliferation genes like Mki67 being highly expressed. Additionally, several candidate genes were identified as potentially crucial in the conversion process. Astrocyte 3 is considered a unique subtype population of astrocytes. CONCLUSIONS Our investigation underscores the diversity of primary neonatal cortical astrocytes and provides critical insights into the potential for astrocyte-to-neuron conversion, which may be harnessed to enhance the efficiency of this astrocyte-neuron conversion process.
Collapse
Affiliation(s)
- Jiaxue Cha
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Peng Zeng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hui Zong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiayi Zhao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiayao Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Haowei Zuo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bowen Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Changjie Shi
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jing Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiuhong Hua
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zixin Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Ru Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Yuan J, Yin C, Peng H, Fang G, Mo B, Qin X, Chen Y, Wang Z, Yu Y, Wang Y, Wang Q. NDRG1 Regulates Iron Metabolism and Inhibits Pathologic Cardiac Hypertrophy. Can J Cardiol 2025; 41:224-240. [PMID: 39427843 DOI: 10.1016/j.cjca.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/26/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Cardiac pathologic hypertrophy, a pathologic physiological alteration in many cardiovascular diseases, can progress to heart failure. The cellular biology underlying myocardial hypertrophy remains to be fully elucidated. Although N-myc downstream-regulated gene 1 (NDRG1) has been reported to participate in cellular proliferation, differentiation, and cellular stress responses, its role in cardiac diseases remains unexplored. Here, we investigated the role of NDRG1 in pathologic hypertrophy. METHOD Cardiomyocyte-specific NDRG1 knockout (KO) transgenic mice and NDRG1-AAV9 were used in mice. Angiotensin II (AngII) stimulation was applied to induce hypertrophy. Histologic, molecular, and RNA-sequencing analyses were performed, and ferroptosis markers and iron levels were studied. We used co-immunoprecipitation (Co-IP) and application of iron chelator to further studied the mechanisms of NDRG1 in cardiac hypertrophy. RESULTS We found that NDRG1 expression is decreased in pathologic hypertrophy induced by AngII stimulation. Conditional KO of NDRG1 in mouse cardiomyocytes led to progressive cardiac hypertrophy and heart failure. Cardiomyocyte-specific overexpression of NDRG1 via AAV9 significantly reversed AngII-induced ventricular hypertrophy and fibrosis. Mechanistically, NDRG1-deficient cardiomyocytes exhibited iron overload and increased ferroptosis, accompanied by elevated levels of reactive oxygen species (ROS) and lipid peroxidation. Subsequently, we confirmed the involvement of NDRG1 in regulating ferroptosis and iron metabolism in myocardial cells. Finally, we identified an interaction between NDRG1 and transferrin in cells. The iron chelator Dp44mT effectively reduced myocardial iron overload and ventricular remodelling induced by NDRG1 deficiency. CONCLUSIONS These findings highlight critical role of NDRG1 in iron metabolism and ferroptosis in cardiomyocytes, suggesting that NDRG1 or iron metabolism may serve as therapeutic targets for cardiac hypertrophy.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chengye Yin
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Peng
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guojian Fang
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Binfeng Mo
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiji Qin
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuhan Chen
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengshuai Wang
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yichi Yu
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qunshan Wang
- Department of Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Xin Y, Zhou S, Chu T, Zhou Y, Xu A. Protective Role of Electroacupuncture Against Cognitive Impairment in Neurological Diseases. Curr Neuropharmacol 2025; 23:145-171. [PMID: 38379403 PMCID: PMC11793074 DOI: 10.2174/1570159x22999240209102116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 02/22/2024] Open
Abstract
Many neurological diseases can lead to cognitive impairment in patients, which includes dementia and mild cognitive impairment and thus create a heavy burden both to their families and public health. Due to the limited effectiveness of medications in treating cognitive impairment, it is imperative to develop alternative treatments. Electroacupuncture (EA), a required method for Traditional Chinese Medicine, has the potential treatment of cognitive impairment. However, the molecular mechanisms involved have not been fully elucidated. Considering the current research status, preclinical literature published within the ten years until October 2022 was systematically searched through PubMed, Web of Science, MEDLINE, Ovid, and Embase. By reading the titles and abstracts, a total of 56 studies were initially included. It is concluded that EA can effectively ameliorate cognitive impairment in preclinical research of neurological diseases and induce potentially beneficial changes in molecular pathways, including Alzheimer's disease, vascular cognitive impairment, chronic pain, and Parkinson's disease. Moreover, EA exerts beneficial effects through the same or diverse mechanisms for different disease types, including but not limited to neuroinflammation, neuronal apoptosis, neurogenesis, synaptic plasticity, and autophagy. However, these findings raise further questions that need to be elucidated. Overall, EA therapy for cognitive impairment is an area with great promise, even though more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Yueyang Xin
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Chu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqun Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aijun Xu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Liu M, Chen J, Sun M, Zhang L, Yu Y, Mi W, Ma Y, Wang G. Protection of Ndrg2 deficiency on renal ischemia-reperfusion injury via activating PINK1/Parkin-mediated mitophagy. Chin Med J (Engl) 2024; 137:2603-2614. [PMID: 38407220 PMCID: PMC11556958 DOI: 10.1097/cm9.0000000000002957] [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: 11/04/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Renal ischemia-reperfusion (R-I/R) injury is the most prevalent cause of acute kidney injury, with high mortality and poor prognosis. However, the underlying pathological mechanisms are not yet fully understood. Therefore, this study aimed to investigate the role of N-myc downstream-regulated gene 2 ( Ndrg2 ) in R-I/R injury. METHODS We examined the expression of Ndrg2 in the kidney under normal physiological conditions and after R-I/R injury by immunofluorescence staining, real-time polymerase chain reaction, and western blotting. We then detected R-I/R injury in Ndrg2-deficient ( Ndrg2-/- ) mice and wild type ( Ndrg2+/+ ) littermates in vivo , and detected oxygen and glucose deprivation and reperfusion (OGD-R) injury in HK-2 cells. We further conducted transcriptomic sequencing to investigate the role of Ndrg2 in R-I/R injury and detected levels of oxidative stress and mitochondrial damage by dihydroethidium staining, biochemical assays, and western blot. Finally, we measured the levels of mitophagy in Ndrg2+/+ and Ndrg2-/- mice after R-I/R injury or HK-2 cells in OGD-R injury. RESULTS Ndrg2 was primarily expressed in renal proximal tubules and its expression was significantly decreased 24 h after R-I/R injury. Ndrg2-/- mice exhibited significantly attenuated R-I/R injury compared to Ndrg2+/+ mice. Transcriptomics profiling showed that Ndrg2 deficiency induced perturbations of multiple signaling pathways, downregulated inflammatory responses and oxidative stress, and increased autophagy following R-I/R injury. Further studies revealed that Ndrg2 deficiency reduced oxidative stress and mitochondrial damage. Notably, Ndrg2 deficiency significantly activated phosphatase and tensin homologue on chromosome ten-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy. The downregulation of NDRG2 expression significantly increased cell viability after OGD-R injury, increased the expression of heme oxygenase-1, decreased the expression of nicotinamide adenine dinucleotide phosphate oxidase 4, and increased the expression of the PINK1/Parkin pathway. CONCLUSION Ndrg2 deficiency might become a therapy target for R-I/R injury by decreasing oxidative stress, maintaining mitochondrial homeostasis, and activating PINK1/Parkin-mediated mitophagy.
Collapse
Affiliation(s)
- Min Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jianwen Chen
- Department of Nephrology, The First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Miao Sun
- Department of Anesthesiology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Lixia Zhang
- Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Yao Yu
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Weidong Mi
- Department of Anesthesiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
5
|
Wu L, Cheng Y, Wang R, Sun S, Ma B, Zhang Z. NDRG2 regulates glucose metabolism and ferroptosis of OGD/R-treated astrocytes by the Wnt/β-catenin signaling. J Biochem Mol Toxicol 2024; 38:e23827. [PMID: 39193856 DOI: 10.1002/jbt.23827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 07/10/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Ischemic stroke is one main type of cerebrovascular disorders with leading cause of death and disability worldwide. Astrocytes are the only nerve cell type storing glycogen in the brain, which regulate the glucose metabolism and handle the energy supply and survive of neurons. Astrocyte ferroptosis contributes to neuron injury in brain disorders. N-myc downstream-regulated gene 2 (NDRG2) has been implicated in the progression of brain diseases, including ischemic stroke. However, whether NDRG2 could affect the glucose metabolism and ferroptosis of astrocytes during ischemic stroke remains largely unknown. Mouse astrocytes were treated with oxygen-glucose deprivation/reoxygenation (OGD/R) to establish the in vitro model. Glial fibrillary acidic protein, NDRG2, Wnt3a and β-catenin expression levels were detected by immunofluorescence staining and western blot analyses. Glucose metabolism was investigated by glucose uptake, lactate production, nicotinamide adenine dinucleotide phosphate hydrogen/nicotinamide adenine dinucleotide phosphate (NADPH/NADP+), ATP and glycolysis enzymes (HK2, PKM2 and lactate dehydrogenase A [LDHA]) levels. Ferroptosis was assessed via reactive oxygen species (ROS), glutathione (GSH), iron and ferroptosis-related markers (GPX4 and PTGS2) contents. Glycolysis enzymes and ferroptosis-related markers levels were measured via western blot. NDRG2 expression was elevated in OGD/R-induced astrocytes. NDRG2 overexpression aggravated OGD/R-induced loss of glucose metabolism through reducing glucose uptake, lactate production, NADPH/NADP+ and ATP levels. NDRG2 upregulation exacerbated OGD/R-caused reduction of glycolysis enzymes (HK2, PKM2 and LDHA) levels. NDRG2 promoted OGD/R-induced ferroptosis of astrocytes by increasing ROS, iron and PTGS2 levels and decreasing GSH and GPX4 levels. NDRG2 overexpression enhanced OGD/R-induced decrease of Wnt/β-catenin signaling activation by reducing Wnt3a and β-catenin expression. NDRG2 silencing played an opposite effect. Inhibition of Wnt/β-catenin signaling activation by IWR-1 attenuated the influences of NDRG2 knockdown on glucose metabolism, glycolysis enzymes levels and ferroptosis. These findings demonstrated that NDRG2 contributes to OGD/R-induced inhibition of glucose metabolism and promotion of ferroptosis in astrocytes through inhibiting Wnt/β-catenin signaling activation, which might be associated with ischemic stroke progression.
Collapse
Affiliation(s)
- Lin Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yingying Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Runfeng Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shukai Sun
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bo Ma
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhiguo Zhang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
Zhang L, Gu W, Liu T, Pei H, Ma Y, Zhao Y, Huang S, Chen M. NDRG2 Deficiency Exacerbates UVB-Induced Skin Inflammation and Oxidative Stress Damage. Inflammation 2024:10.1007/s10753-024-02121-3. [PMID: 39145786 DOI: 10.1007/s10753-024-02121-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/16/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
UVB radiation induces inflammatory and oxidative stress responses, contributing to skin damage, yet the underlying mechanisms are not fully understood. N-Myc downstream-regulated gene 2 (NDRG2), an emerging stress-associated gene, remains unexplored in UVB-induced skin injury. In this study, we detected skin NDRG2 expression after UVB irradiation for the first time and further used Ndrg2 knockout mice to clarify the role of NDRG2 in UVB-induced skin injury. Three-month-old male Ndrg2+/+ and Ndrg2-/- mice (16-18g) were exposed to UVB to induce acute skin damage, and then dorsal skin samples were collected for subsequent analyses. UVB-induced skin damage was scored. Western Blot Analysis, immunofluorescence (IF) double labeling, and immunohistochemistry (IHC) were employed to assess NDRG2 expression and/or distribution. The concentrations of TNF-α, IL-6, IL-1β, MPO, MMP8, superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) were quantitatively assessed using enzyme-linked immunosorbent assay (ELISA). Hematoxylin and eosin (HE) staining were employed to determine pathological changes. RNA sequencing and analysis were performed to estimate transcript expression levels and analyze mRNA expression. DESeq2 software was employed to identify differentially expressed genes (DEGs). DEGs were visualized using volcanic and heat maps. Gene Ontology (GO) functions and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were analyzed to identify primary biological functions, metabolic pathways, or signal transduction pathways associated with DEGs. UVB-challenged Ndrg2-/- mice exhibited significantly exacerbated skin damage (erythema, edema, and erosion), neutrophil infiltration, and apoptosis compared to Ndrg2+/+ mice. Furthermore, UVB-challenged Ndrg2-/- mice displayed significantly elevated pro-inflammatory cytokines, myeloperoxidase (MPO), matrix metalloproteinase-8 (MMP8), and reduced antioxidant expression. RNA sequencing identified 1091 significantly differentially expressed genes enriched in inflammation, immune response, and oxidative stress pathways. In conclusion, the deficiency of Ndrg2 markedly exacerbated UVB-induced skin damage by promoting inflammatory responses and inhibiting antioxidant responses. This suggests that stabilizing NDRG2 expression holds promise as a therapeutic strategy for protecting against UVB-induced skin damage.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Plastic and Reconstructive Surgery, Senior Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese, PLA General Hospital and PLA Medical College, Beijing, 100048, China
| | - Weijie Gu
- Department of Dermatology, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Tian Liu
- Senior Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital and PLA Medical College, Beijing, 100048, China
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, 510010, China
| | - Haina Pei
- Department of Plastic and Reconstructive Surgery, Senior Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese, PLA General Hospital and PLA Medical College, Beijing, 100048, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Yi Zhao
- Department of Dermatology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China.
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital, PLA Medical College, Beijing, 100853, China.
| | - Minliang Chen
- Department of Plastic and Reconstructive Surgery, Senior Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese, PLA General Hospital and PLA Medical College, Beijing, 100048, China.
| |
Collapse
|
7
|
Long CM, Li Z, Song W, Zeng X, Yang R, Lu L. The Roles of Non-coding RNA Targeting Astrocytes in Cerebral Ischemia. Mol Neurobiol 2024; 61:5814-5825. [PMID: 38236344 DOI: 10.1007/s12035-023-03898-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024]
Abstract
Astrocytes are key targets for treating cerebral ischemia in the central nervous system. Non-coding RNAs (ncRNAs) participate in the pathological processes of astrocytes in cerebral ischemia. Recent reports suggest that ncRNAs ameliorate the outcome of cerebral ischemia by mediating astrocytes' inflammatory reaction, oxidative stress, excitotoxicity, autophagy, and apoptosis. Reconstructing cellular systems might offer a promising strategy for treating cerebral ischemia. This review briefly discusses the potential of ncRNAs as drug targets and explores the molecular regulatory mechanisms through which ncRNAs target astrocytes in cerebral ischemia. It provides an overview of the current research, discusses ncRNAs' implications as clinical markers for cerebral ischemia, and anticipates that ongoing research on ncRNAs may contribute to novel therapeutic approaches for treating this condition.
Collapse
Affiliation(s)
- Chun-Mei Long
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Zhen Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Wang Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Xin Zeng
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China
| | - Rui Yang
- The Endocrinology Department, Lanzhou Hospital of Traditional Chinese Medicine, Lanzhou, 73000, Gansu, China
| | - Li Lu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 73000, Gansu, China.
- Medical College of Lanzhou University, 199 Dong gang West Road, Cheng guan District, Lanzhou, China.
| |
Collapse
|
8
|
Jiang S, Zhu G, Tan Y, Zhou T, Zheng S, Wang F, Lei W, Liu X, Du J, Tian M. Identification of VEGFs-related gene signature for predicting microangiogenesis and hepatocellular carcinoma prognosis. Aging (Albany NY) 2024; 16:10321-10347. [PMID: 38874512 PMCID: PMC11236318 DOI: 10.18632/aging.205931] [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: 08/25/2023] [Accepted: 04/08/2024] [Indexed: 06/15/2024]
Abstract
Microangiogenesis is an important prognostic factor in various cancers, including hepatocellular carcinoma (HCC). The Vascular Endothelial Growth Factor (VEGF) has been shown to contribute to tumor angiogenesis. Recently, several studies have investigated the regulation of VEGF production by a single gene, with few researchers exploring all genes that affect VEGF production. In this study, we comprehensively analyzed all genes affecting VEGF production in HCC and developed a risk model and gene-based risk score based on VEGF production. Moreover, the model's predictive capacity on prognosis of HCCs was verified using training and validation datasets. The developed model showed good prediction of the overall survival rate. Patients with a higher risk score experienced poor outcomes compared to those with a lower risk score. Furthermore, we identified the immunological causes of the poor prognosis of patients with high-risk scores comparing with those with low-risk scores.
Collapse
Affiliation(s)
- Shengpan Jiang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Guoting Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yiqing Tan
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Tao Zhou
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Shilin Zheng
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Fuhua Wang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Wenfeng Lei
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Xuan Liu
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Jinjun Du
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| | - Manman Tian
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| |
Collapse
|
9
|
Jiang T, Li Y, He S, Huang N, Du M, Zhai Q, Pu K, Wu M, Yan C, Ma Z, Wang Q. Reprogramming astrocytic NDRG2/NF-κB/C3 signaling restores the diabetes-associated cognitive dysfunction. EBioMedicine 2023; 93:104653. [PMID: 37329577 DOI: 10.1016/j.ebiom.2023.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 04/14/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Dementia is a serious complication in patients with diabetes-associated cognitive dysfunction (DACD). In this study, we aim to explore the protective effect of exercise on DACD in diabetic mice, and the role of NDRG2 as a potential guarder for reversing the pathological structure of neuronal synapses. METHODS Seven weeks of standardized exercise at moderate intensity was carried out using an animal treadmill in the vehicle + Run and STZ + Run groups. Based on quantitative transcriptome and tandem mass tag (TMT) proteome sequencing, weighted gene co-expression analysis (WGCNA) and gene set enrichment analysis (GSEA) were used to investigate the activation of complement cascades to injury neuronal synaptic plasticity. Golgi staining, Western blotting, immunofluorescence staining, and electrophysiology were used to verify the reliability of sequencing data. The role of NDRG2 was assessed by overexpressing or inhibiting the NDRG2 gene in vivo. Moreover, we estimated the cognitive function in diabetic or normal patients using DSST scores. FINDINGS Exercise reversed the injury of neuronal synaptic plasticity and the downregulation of astrocytic NDRG2 in diabetic mice, which succeeded in attenuating DACD. The deficiency of NDRG2 aggravated the activation of complement C3 by accelerating the phosphorylation of NF-κB, ultimately leading to synaptic injury and cognitive dysfunction. Conversely, the overexpression of NDRG2 promoted astrocytic remodeling by inhibiting complement C3, thus attenuating synaptic injury and cognitive dysfunction. Meanwhile, C3aR blockade rescued dendritic spines loss and cognitive deficits in diabetic mice. Moreover, the average DSST score of diabetic patients was significantly lower than that of non-diabetic peers. Levels of complement C3 in human serum were elevated in diabetic patients compared to those in non-diabetic patients. INTERPRETATION Our findings illustrate the effectiveness and integrative mechanism of NDRG2-induced improvement of cognition from a multi-omics perspective. Additionally, they confirm that the expression of NDRG2 is closely related to cognitive function in diabetic mice and the activation of complement cascades accelerated impairment of neuronal synaptic plasticity. NDRG2 acts as a regulator of astrocytic-neuronal interaction via NF-κB/C3/C3aR signaling to restore synaptic function in diabetic mice. FUNDING This study was supported by the National Natural Science Foundation of China (No. 81974540, 81801899, 81971290), the Key Research and Development Program of Shaanxi (Program No. 2022ZDLSF02-09) and Fundamental Research Funds for the Central Universities (Grant No. xzy022019020).
Collapse
Affiliation(s)
- Tao Jiang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Shuxuan He
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ning Huang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qian Zhai
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Kairui Pu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Meiyan Wu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhi Ma
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
10
|
Wang C, Wang X, Zheng H, Yao J, Xiang Y, Liu D. The ndrg2 Gene Regulates Hair Cell Morphogenesis and Auditory Function during Zebrafish Development. Int J Mol Sci 2023; 24:10002. [PMID: 37373150 PMCID: PMC10297845 DOI: 10.3390/ijms241210002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Damages of sensory hair cells (HCs) are mainly responsible for sensorineural hearing loss, however, its pathological mechanism is not yet fully understood due to the fact that many potential deafness genes remain unidentified. N-myc downstream-regulated gene 2 (ndrg2) is commonly regarded as a tumor suppressor and a cell stress-responsive gene extensively involved in cell proliferation, differentiation, apoptosis and invasion, while its roles in zebrafish HC morphogenesis and hearing remains unclear. Results of this study suggested that ndrg2 was highly expressed in the HCs of the otic vesicle and neuromasts via in situ hybridization and single-cell RNA sequencing. Ndrg2 loss-of-function larvae showed decreased crista HCs, shortened cilia, and reduced neuromasts and functional HCs, which could be rescued by the microinjection of ndrg2 mRNA. Moreover, ndrg2 deficiency induced attenuated startle response behaviors to sound vibration stimuli. Mechanistically, there were no detectable HC apoptosis and supporting cell changes in the ndrg2 mutants, and HCs were capable of recovering by blocking the Notch signaling pathway, suggesting that ndrg2 was implicated in HC differentiation mediated by Notch. Overall, our study demonstrates that ndrg2 plays crucial roles in HC development and auditory sensory function utilizing the zebrafish model, which provides new insights into the identification of potential deafness genes and regulation mechanism of HC development.
Collapse
Affiliation(s)
- Cheng Wang
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (C.W.); (J.Y.); (Y.X.)
| | - Xin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226001, China;
| | - Hao Zheng
- School of Medicine, Nantong University, Nantong 226001, China;
| | - Jia Yao
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (C.W.); (J.Y.); (Y.X.)
| | - Yuqing Xiang
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (C.W.); (J.Y.); (Y.X.)
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (C.W.); (J.Y.); (Y.X.)
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226001, China;
| |
Collapse
|
11
|
Zhao Y, Fan X, Wang Q, Zhen J, Li X, Zhou P, Lang Y, Sheng Q, Zhang T, Huang T, Zhao Y, Lv Z, Wang R. ROS promote hyper-methylation of NDRG2 promoters in a DNMTS-dependent manner: Contributes to the progression of renal fibrosis. Redox Biol 2023; 62:102674. [PMID: 36989575 PMCID: PMC10074964 DOI: 10.1016/j.redox.2023.102674] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Renal fibrosis is the common histopathological feature of chronic kidney diseases (CKD), and there is increasing evidence that epigenetic regulation is involved in the occurrence and progression of renal fibrosis. N-myc downstream-regulated gene 2 (NDRG2) is significantly down-regulated in renal fibrosis, the mechanism of which remains unclear. Previous studies have confirmed that the inhibition of NDRG2 expression in tumor cells is related to hyper-methylation, mainly regulated by DNA methyltransferases (DNMTS). Herein, we explored the expression of NDRG2 and its epigenetic regulatory mechanism in renal fibrosis. The results showed that the expression of NDRG2 was significantly inhibited in vivo and in vitro, while the overexpression of NDRG2 effectively alleviated renal fibrosis. Meanwhile, we found that the expression of DNMT1/3A/3B was significantly increased in hypoxia-induced HK2 cells and Unilateral Ureteral Obstruction (UUO) mice accompanied by hyper-methylation of the NDGR2 promoter. Methyltransferase inhibitor (5-AZA-dC) corrected the abnormal expression of DNMT1/3A/3B, reduced the methylation level of NDRG2 promoter and restored the expression of NDRG2. The upstream events that mediate changes in NDRG2 methylation were further explored. Reactive oxygen species (ROS) are important epigenetic regulators and have been shown to play a key role in renal injury due to various causes. Accordingly, we further explored whether ROS could induce DNA-epigenetic changes of the expression of NDRG2 and then participated in the development of renal fibrosis. Our results showed that mitochondria-targeted antioxidants (Mito-TEMPO) could reverse the epigenetic inhibition of NDRG2 in a DNMT-sensitive manner, showing strong ability of DNA demethylation, exhibiting epigenetic regulation and anti-fibrosis effects similar to 5-AZA-dC. More importantly, the anti-fibrotic effects of 5-AZA-dC and Mito-TEMPO were eliminated in HK2 cells with NDRG2 knockdown. These findings highlight that targeting ROS-mediated hyper-methylation of NDRG2 promoter is a potentially effective therapeutic strategy for renal fibrosis, which will provide new insights into the treatment of CKD.
Collapse
|
12
|
Inhibition of PI3K/Akt/mTOR signaling by NDRG2 contributes to neuronal apoptosis and autophagy in ischemic stroke. J Stroke Cerebrovasc Dis 2023; 32:106984. [PMID: 36652790 DOI: 10.1016/j.jstrokecerebrovasdis.2023.106984] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Astrocytic N-myc downstream-regulated gene 2 (NDRG2), a differentiation- and stress-associated molecule, has been involved in the cause of ischemic stroke (IS). However, its downstream effector in IS remains unclear. This study aimed to characterize expression of NDRG2 in IS patients and rats and to investigate the underlying mechanism. METHODS The protein expression of NDRG2 and mammalian target of the rapamycin (mTOR) and the extent of mTOR phosphorylation in plasma of IS patients were detected by ELISA. An oxygen-glucose deprivation model was established in mouse neuronal cells CATH.a, followed by cell counting kit-8, flow cytometry, TUNEL, and western blot assays to examine cell viability, apoptosis and autophagy. Finally, the effect of NDRG2-mediated phosphatidylinositol 3-kinase/protein kinase-B/mTOR (PI3K/AKT/mTOR) pathway on neuronal apoptosis and autophagy was verified in rats treated with middle cerebral artery occlusion. RESULTS NDRG2 was highly expressed in the plasma of IS patients, while the extent of mTOR phosphorylation was reduced in IS patients. NDRG2 blocked the PI3K/Akt/mTOR signaling through dephosphorylation. Depletion of NDRG2 suppressed apoptosis and autophagy in CATH.a cells, which was reversed by a dual inhibitor of PI3K and mTOR, BEZ235. In vivo experiments confirmed that NDRG2 promoted neuronal apoptosis and autophagy by dephosphorylating and blocking the PI3K/Akt/mTOR signaling. CONCLUSION The present study has shown that NDRG2 impairs the PI3K/Akt/mTOR pathway via dephosphorylation to promote neuronal apoptosis and autophagy in IS. These findings provide potential targets for future clinical therapies for IS.
Collapse
|
13
|
Feng D, Zhou J, Liu H, Wu X, Li F, Zhao J, Zhang Y, Wang L, Chao M, Wang Q, Qin H, Ge S, Liu Q, Zhang J, Qu Y. Astrocytic NDRG2-PPM1A interaction exacerbates blood-brain barrier disruption after subarachnoid hemorrhage. SCIENCE ADVANCES 2022; 8:eabq2423. [PMID: 36179025 PMCID: PMC9524825 DOI: 10.1126/sciadv.abq2423] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/15/2022] [Indexed: 06/01/2023]
Abstract
Blood-brain barrier (BBB) injury critically exacerbates the poor prognosis of patients with subarachnoid hemorrhage (SAH). The massively increased matrix metalloproteinases 9 (MMP-9) plays a deleterious role in BBB. However, the main source and mechanism of MMP-9 production after SAH remain unclear. We reported that the increased MMP-9 was mainly derived from reactive astrocytes after SAH. Ndrg2 knockout in astrocytes inhibited MMP-9 expression after SAH and attenuated BBB damage. Astrocytic Ndrg2 knockout decreased the phosphorylation of Smad2/3 and the transcription of MMP-9. Notably, cytoplasmic NDRG2 bound to the protein phosphatase PPM1A and restricted the dephosphorylation of Smad2/3. Accordingly, TAT-QFNP12, a novel engineered peptide that could block the NDRG2-PPM1A binding and reduce Smad2/3 dephosphorylation, decreased astrocytic MMP-9 production and BBB disruption after SAH. In conclusion, this study identified NDRG2-PPM1A signaling in reactive astrocytes as a key switch for MMP-9 production and provided a novel therapeutic avenue for BBB protection after SAH.
Collapse
Affiliation(s)
- Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Jinpeng Zhou
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Fei Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Junlong Zhao
- Department of Medical Genetics and Development Biology, Fourth Military Medical University, Xi’an 710032, China
| | - Yu Zhang
- Department of Biological Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Lei Wang
- Department of Biological Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Huaizhou Qin
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jian Zhang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an 710032, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| |
Collapse
|
14
|
Lamoureux L, Sajesh B, Slota JA, Medina SJ, Mayor M, Frost KL, Warner B, Manguiat K, Wood H, Kobasa D, Booth SA. Non-Productive Infection of Glial Cells with SARS-CoV-2 in Hamster Organotypic Cerebellar Slice Cultures. Viruses 2022; 14:1218. [PMID: 35746689 PMCID: PMC9227386 DOI: 10.3390/v14061218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
The numerous neurological syndromes associated with COVID-19 implicate an effect of viral pathogenesis on neuronal function, yet reports of direct SARS-CoV-2 infection in the brain are conflicting. We used a well-established organotypic brain slice culture to determine the permissivity of hamster brain tissues to SARS-CoV-2 infection. We found levels of live virus waned after inoculation and observed no evidence of cell-to-cell spread, indicating that SARS-CoV-2 infection was non-productive. Nonetheless, we identified a small number of infected cells with glial phenotypes; however, no evidence of viral infection or replication was observed in neurons. Our data corroborate several clinical studies that have assessed patients with COVID-19 and their association with neurological involvement.
Collapse
Affiliation(s)
- Lise Lamoureux
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Babu Sajesh
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Jessy A. Slota
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, 730 William Ave., Winnipeg, MB R3E 0W3, Canada;
| | - Sarah J. Medina
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Matthew Mayor
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Kathy L. Frost
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Bryce Warner
- Special Pathogens, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada;
| | - Kathy Manguiat
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Heidi Wood
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
| | - Darwyn Kobasa
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, 730 William Ave., Winnipeg, MB R3E 0W3, Canada;
- Special Pathogens, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada;
| | - Stephanie A. Booth
- One Health Division, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington St., Winnipeg, MB R3E 3R2, Canada; (L.L.); (B.S.); (J.A.S.); (S.J.M.); (M.M.); (K.L.F.); (K.M.); (H.W.)
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, 730 William Ave., Winnipeg, MB R3E 0W3, Canada;
| |
Collapse
|
15
|
N-myc Downstream-Regulated Gene 2 (Ndrg2): A Critical Mediator of Estrogen-Induced Neuroprotection Against Cerebral Ischemic Injury. Mol Neurobiol 2022; 59:4793-4804. [PMID: 35622273 DOI: 10.1007/s12035-022-02877-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Abstract
Growing evidence indicates that estrogen plays a pivotal role in neuroprotection against cerebral ischemia, but the molecular mechanism of this protection is still elusive. N-myc downstream-regulated gene 2 (Ndrg2), an estrogen-targeted gene, has been shown to exert neuroprotective effects against cerebral ischemia in male mice. However, the role of Ndrg2 in the neuroprotective effect of estrogen remains unknown. In this study, we first detected NDRG2 expression levels in the cortex and striatum in both female and male mice with western blot analyses. We then detected cerebral ischemic injury by constructing middle cerebral artery occlusion and reperfusion (MCAO-R) models in Ndrg2 knockout or conditional knockdown female mice. We further implemented estrogen, ERα, or ERβ agonist replacement in the ovariectomized (OVX) Ndrg2 knockout or conditional knockdown female mice, then tested for NDRG2 expression, glial fibrillary acidic protein (GFAP) expression, and extent of cerebral ischemic injury. We found that NDRG2 expression was significantly higher in female than in male mice in both the cortex and striatum. Ndrg2 knockouts and conditional knockdowns showed significantly aggravated cerebral ischemic injury in female mice. Estrogen and ERβ replacement treatment (DPN) led to NDRG2 upregulation in both the cortex and striatum of OVX mice. Estrogen and DPN also led to GFAP upregulation in OVX mice. However, the effect of estrogen and DPN in activating astrocytes was lost in Ndrg2 knockout OVX mice and primary cultured astrocytes, but partially retained in conditional knockdown OVX mice. Most importantly, we found that the neuroprotective effects of E2 and DPN against cerebral ischemic injury were lost in Ndrg2 knockout OVX mice but partially retained in conditional knockdown OVX mice. These findings demonstrate that estrogen alleviated cerebral ischemic injury via ERβ upregulation of Ndrg2, which could activate astrocytes, indicating that Ndrg2 is a critical mediator of E2-induced neuroprotection against cerebral ischemic injury.
Collapse
|
16
|
Gao L, Wang C, Qin B, Li T, Xu W, Lenahan C, Ying G, Li J, Zhao T, Zhu Y, Chen G. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase Suppresses Neuronal Apoptosis by Increasing Glycolysis and "cyclin-dependent kinase 1-Mediated Phosphorylation of p27 After Traumatic Spinal Cord Injury in Rats. Cell Transplant 2021; 29:963689720950226. [PMID: 32841050 PMCID: PMC7563815 DOI: 10.1177/0963689720950226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Apoptosis is a vital pathological factor that accounts for the poor prognosis of
traumatic spinal cord injury (t-SCI). The
6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3) is a critical
regulator for energy metabolism and proven to have antiapoptotic effects. This
study aimed to investigate the neuroprotective role of PFKFB3 in t-SCI. A
compressive clip was introduced to establish the t-SCI model. Herein, we
identified that PFKFB3 was extensively distributed in neurons, and PFKFB3 levels
significantly increased and peaked 24 h after t-SCI. Additionally, knockdown of
PFKFB3 inhibited glycolysis, accompanied by aggravated neuronal apoptosis and
white matter injury, while pharmacological activation of PFKFB3 with meclizine
significantly enhanced glycolysis, attenuated t-SCI-induced spinal cord injury,
and alleviated neurological impairment. The PFKFB3 agonist, meclizine, activated
cyclin-dependent kinase 1 (CDK1) and promoted the phosphorylation of p27,
ultimately suppressing neuronal apoptosis. However, the neuroprotective effects
of meclizine against t-SCI were abolished by the CDK1 antagonist, RO3306. In
summary, our data demonstrated that PFKFB3 contributes robust neuroprotection
against t-SCI by enhancing glycolysis and modulating CDK1-related antiapoptotic
signals. Moreover, targeting PFKFB3 may be a novel and promising therapeutic
strategy for t-SCI.
Collapse
Affiliation(s)
- Liansheng Gao
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chun Wang
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bing Qin
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tao Li
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weilin Xu
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cameron Lenahan
- 448838Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Guangyu Ying
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianru Li
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tengfei Zhao
- Department of Orthopedics, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongjian Zhu
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Gao Chen
- Department of Neurosurgery, 89681Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Ravi K, Paidas MJ, Saad A, Jayakumar AR. Astrocytes in rare neurological conditions: Morphological and functional considerations. J Comp Neurol 2021; 529:2676-2705. [PMID: 33496339 DOI: 10.1002/cne.25118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 01/06/2023]
Abstract
Astrocytes are a population of central nervous system (CNS) cells with distinctive morphological and functional characteristics that differ within specific areas of the brain and are widely distributed throughout the CNS. There are mainly two types of astrocytes, protoplasmic and fibrous, which differ in morphologic appearance and location. Astrocytes are important cells of the CNS that not only provide structural support, but also modulate synaptic activity, regulate neuroinflammatory responses, maintain the blood-brain barrier, and supply energy to neurons. As a result, astrocytic disruption can lead to widespread detrimental effects and can contribute to the pathophysiology of several neurological conditions. The characteristics of astrocytes in more common neuropathologies such as Alzheimer's and Parkinson's disease have significantly been described and continue to be widely studied. However, there still exist numerous rare neurological conditions in which astrocytic involvement is unknown and needs to be explored. Accordingly, this review will summarize functional and morphological changes of astrocytes in various rare neurological conditions based on current knowledge thus far and highlight remaining neuropathologies where astrocytic involvement has yet to be investigated.
Collapse
Affiliation(s)
- Karthik Ravi
- University of Michigan, Ann Arbor, Michigan, USA
| | - Michael J Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA
| | - Ali Saad
- Pathology and Laboratory Medicine, University of Miami School of Medicine, Miami, Florida, USA
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA.,South Florida VA Foundation for Research and Education Inc, Miami, Florida, USA.,General Medical Research Neuropathology Section, R&D Service, Veterans Affairs Medical Centre, Miami, Florida, USA
| |
Collapse
|
18
|
Takarada-Iemata M, Yoshihara T, Okitani N, Iwata K, Hattori T, Ishii H, Roboon J, Nguyen DT, Fan Q, Tamatani T, Nishiuchi T, Asano M, Hori O. Abnormal social behavior and altered gene expression in mice lacking NDRG2. Neurosci Lett 2020; 743:135563. [PMID: 33359046 DOI: 10.1016/j.neulet.2020.135563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
N-myc downstream-regulated gene 2 (NDRG2), a member of the NDRG family, has multiple functions in cell proliferation, differentiation, and stress responses, and is predominantly expressed by astrocytes in the central nervous system. Previous studies including ours demonstrated that NDRG2 is involved in various central nervous system pathologies. However, the significance of NDRG2 in neurodevelopment is not fully understood. Here, we investigated the expression profile of NDRG2 during postnatal brain development, the role of NDRG2 in social behavior, and transcriptome changes in the brain of NDRG2-deficient mice. NDRG2 expression in the brain increased over time from postnatal day 1 to adulthood. Deletion of NDRG2 resulted in abnormal social behavior, as indicated by reduced exploratory activity toward a novel mouse in a three-chamber social interaction test. Microarray analysis identified genes differentially expressed in the NDRG2-deficient brain, and upregulated gene expression of Bmp4 and Per2 was confirmed by quantitative PCR analysis. Expression of both these genes and the encoded proteins increased over time during postnatal brain development, similar to NDRG2. Gene expression of Bmp4 and Per2 was upregulated in cultured astrocytes isolated from NDRG2-deficient mice. These results suggest that NDRG2 contributes to brain development required for proper social behavior by modulating gene expression in astrocytes.
Collapse
Affiliation(s)
- Mika Takarada-Iemata
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan.
| | - Toru Yoshihara
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nahoko Okitani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Yoshida-gun, Fukui, 910-1193, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Dinh Thi Nguyen
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Qiyan Fan
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takumi Nishiuchi
- Institute for Gene Research, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
19
|
Cheng H, Zhang L, Xia F, Jin L, Liu S, Ren H, Zhu C, Ji Q, Tang J. Astrocytic NDRG2 is critical in the maintenance of neuropathic pain. Brain Behav Immun 2020; 89:300-313. [PMID: 32688030 DOI: 10.1016/j.bbi.2020.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Activation of astrocytes and abnormal synaptic glutamate metabolism are closely associated with the induction and maintenance of neuropathic pain (NP), but the exact mechanism underlying this association remains unclear. N-myc downstream-regulated gene 2 (NDRG2), a novel tumor-suppressor protein and stress-response gene, is involved in the pathogenesis of several neurodegenerative diseases. However, its role in nociceptive transduction has rarely been investigated. Here, we found that NDRG2, which was mainly expressed in the astrocytes in the central nervous system (CNS), was increased in the spinal cord of a spinal nerve ligation (SNL) rat model for NP. Suppression of NDRG2 by intrathecal injection of an NDRG2-RNAi-adenovirus significantly alleviated SNL-induced mechanical and thermal hypersensitivity, as well as elevated astrocytic glutamate transporter 1 (GLT-1) expression and downregulated pro-inflammatory cytokine levels, in the spinal dorsal horn of rats on Day 10 after SNL. Furthermore, in lipopolysaccharide (LPS)-stimulated primary astrocytic cultures derived from neonatal rats, inhibition of NDRG2 significantly reversed both the LPS-induced activation of astrocytes and decreased expression of GLT-1. By contrast, overexpression of NDRG2 by an adenoviral vector carrying NDRG2 resulted in astrocytic activation, aberrant glutamatergic neurotransmission, and spontaneous nociceptive responses in rats. Intrathecal injection of AG490, which is an inhibitor of the Janus tyrosine kinase and signal transducer and activator of the transcription 3 (JAK/STAT3) signaling pathway, significantly attenuated both mechanical and thermal hyperalgesia, as well as inhibited reactive astrocytes and restored normal expression levels of astrocytic GLT-1, in the spinal dorsal horn of NDRG2-overexpression rats. In conclusion, spinal astrocytic NDRG2 is critical in the maintenance of NP. Moreover, NDRG2 modulates astrocytic activation and inflammatory responses via regulating GLT-1 expression through the JAK/STAT3 signaling pathway. Our findings suggested that NDRG2 could be a novel therapeutic target for the treatment of NP.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fei Xia
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Jin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Suting Liu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hongwei Ren
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chao Zhu
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Ji
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Jun Tang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|