1
|
Xu W, Ma X, Zhang C. Analysis of influencing factors for postoperative brain injury in patients with cardiac surgery. Brain Inj 2024:1-10. [PMID: 39642047 DOI: 10.1080/02699052.2024.2430386] [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: 07/23/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024]
Abstract
OBJECTIVE To identify risk factors in people with heart disease prior to undergoing cardiac surgery and to provide a prediction model. METHODS 106 patients who underwent cardiac surgery were selected by convenience sampling method. Case-control study design was used to divide them into control and observation groups according to whether there was postoperative brain injury. General demographic data, perioperative surgery, postoperative recovery, past medical history and preoperative conditions, laboratory test indicators were collected. Univariate and multivariate Logistic regression analyses were performed to select influencing factors. A nomogram model for predicting postoperative brain injury was established. RESULTS Univariate Logistic regression analysis showed that age, female, stroke, diabetes mellitus, perioperative administration of dexmedetomidine, intraoperative use of cardiopulmonary bypass were related factors for postoperative brain injury(p < 0.05). Multivariate Logistic regression analysis showed that females, history of stroke, and intraoperative use of cardiopulmonary bypass were risk factors, and perioperative administration of dexmedetomidine was protective factor(p < 0.05). Nomogram model was established with an AUC of 0.847 and accuracy of 84.7%. CONCLUSION Preoperative stroke history and females are risk factors for brain injury after coronary artery bypass grafting under cardiopulmonary bypass, and perioperative use of dexmedetomidine is protective factor.
Collapse
Affiliation(s)
- Weifang Xu
- The First Affiliated Hospital of XinJiang Medical University, School of Public Health, Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| | - Xiaolan Ma
- School of Nursing, Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| | - Chen Zhang
- School of Public Health, Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| |
Collapse
|
2
|
Szeky B, Jurakova V, Fouskova E, Feher A, Zana M, Karl VR, Farkas J, Bodi-Jakus M, Zapletalova M, Pandey S, Kucera R, Lochman J, Dinnyes A. Efficient derivation of functional astrocytes from human induced pluripotent stem cells (hiPSCs). PLoS One 2024; 19:e0313514. [PMID: 39630626 PMCID: PMC11616838 DOI: 10.1371/journal.pone.0313514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Astrocytes are specialized glial cell types of the central nervous system (CNS) with remarkably high abundance, morphological and functional diversity. Astrocytes maintain neural metabolic support, synapse regulation, blood-brain barrier integrity and immunological homeostasis through intricate interactions with other cells, including neurons, microglia, pericytes and lymphocytes. Due to their extensive intercellular crosstalks, astrocytes are also implicated in the pathogenesis of CNS disorders, such as ALS (amyotrophic lateral sclerosis), Parkinson's disease and Alzheimer's disease. Despite the critical importance of astrocytes in neurodegeneration and neuroinflammation are recognized, the lack of suitable in vitro systems limits their availability for modeling human brain pathologies. Here, we report the time-efficient, reproducible generation of astrocytes from human induced pluripotent stem cells (hiPSCs). Our hiPSC-derived astrocytes expressed characteristic astrocyte markers, such as GFAP, S100b, ALDH1L1 and AQP4. Furthermore, hiPSC-derived astrocytes displayed spontaneous calcium transients and responded to inflammatory stimuli by the secretion of type A1 and type A2 astrocyte-related cytokines.
Collapse
Affiliation(s)
| | - Veronika Jurakova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Eliska Fouskova
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | | | | | | | | | | | - Martina Zapletalova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Shashank Pandey
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Radek Kucera
- Department of Pharmacology and Toxicology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Immunochemistry Diagnostics, University Hospital Pilsen, Pilsen, Czech Republic
| | - Jan Lochman
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Andras Dinnyes
- BioTalentum Ltd, Godollo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Godollo, Hungary
| |
Collapse
|
3
|
Lu L, Ye J, Yi D, Qi T, Luo T, Wu S, Yang L, Li L, Zhang H, Chen D. Runx2 Suppresses Astrocyte Activation and Astroglial Scar Formation After Spinal Cord Injury in Mice. Mol Neurobiol 2024; 61:10820-10829. [PMID: 38789894 PMCID: PMC11584425 DOI: 10.1007/s12035-024-04212-6] [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: 09/28/2023] [Accepted: 03/29/2024] [Indexed: 05/26/2024]
Abstract
After spinal cord injury, astrocytes undergo a reactive process and form an astroglial scar, which impedes the regeneration of axons. The role of Runx2 in promoting the transformation of astrocytes in the central nervous system is well-established. However, it remains unclear whether Runx2 also plays a role in the development of astroglial scar, and the precise underlying mechanism has yet to be identified. Recently, our study using cell culture and animal models has demonstrated that Runx2 actually suppresses astrocyte activation and the formation of astroglial scar following injury. The initial results demonstrated an increase in the expression of Runx2 in astrocytes following in vivo injury. Subsequently, the overexpression of Runx2 resulted in the inhibition of astrocyte activation, reduction in the total area of astroglial scar, and restoration of neural function after 14 days of injury. However, these effects were reversed by CADD522. These findings indicate that Runx2 could potentially serve as a therapeutic intervention for spinal cord injury (SCI). Furthermore, our findings suggest that the Nuclear-matrix-targeting signal (NMTS) of Runx2 is associated with its effect. In summary, the study's results propose that targeting Runx2 may be a promising treatment approach for reactive astrocytes and astroglial scar in the recovery of SCI.
Collapse
Affiliation(s)
- Leilei Lu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jiazong Ye
- Department of Ultrasound, Dongtou District People's Hospital, Wenzhou, Zhejiang, 325700, China
| | - Dafa Yi
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tengfei Qi
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tong Luo
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Silei Wu
- The Wenzhou Third Clinical Institute Affiliated To Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lei Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Daqing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
4
|
Yin S, Xia F, Zou W, Jiang F, Shen K, Sun B, Lu Z. Ginsenoside Rg1 regulates astrocytes to promote angiogenesis in spinal cord injury via the JAK2/STAT3 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118531. [PMID: 38971343 DOI: 10.1016/j.jep.2024.118531] [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: 04/24/2024] [Revised: 06/12/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng (Panax ginseng C. A. Mey) is a common traditional Chinese medicine used for anti-inflammation, anti-apoptosis, anti-oxidative stress, and neuroprotection. Ginsenosides Rg1, the main active components isolated from ginseng, may be a feasible therapy for spinal cord injury (SCI). AIMS OF THE STUDY SCI causes endothelial cell death and blood vessel rupture, ultimately resulting in long-term neurological impairment. As a result, encouraging spinal angiogenesis may be a feasible therapy for SCI. This investigation aimed to validate the capacity of ginsenoside Rg1 in stimulating angiogenesis within the spinal cord. MATERIALS AND METHODS Rats with SCI were injected intraperitoneally with ginsenoside Rg1. The effectiveness of ginsenoside Rg1 was assessed using the motor function score and the motor-evoked potential (MEP). Immunofluorescence techniques were applied to identify the spinal cord's angiogenesis. Angiogenic factors were examined through Western Blot (WB) and Immunohistochemistry. Oxygen-glucose deprivation (OGD) was employed to establish the hypoxia-ischemia model in vitro, and astrocytes (As) were given ginsenoside Rg1 and co-cultured with spinal cord microvascular endothelial cells (SCMECs). Immunofluorescence, wound healing test, and tube formation assay were used to identify the co-cultured SCMECs' activity. Finally, network pharmacology analysis and siRNA transfection were applied to verify the mechanism of ginsenoside Rg1 promoting angiogenesis. RESULTS The rats with SCI treated with ginsenoside Rg1 indicated more significant functional recovery, more pronounced angiogenesis, and higher levels of angiogenic factor expression. In vitro, the co-culture system with ginsenoside Rg1 intervention improved SCMECs' capacity for proliferating, migrating, and forming tubes, possibly by promoting the expression of vascular endothelial growth factor (VEGF) in As via the janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. CONCLUSION Ginsenoside Rg1 can regulate As to promote angiogenesis, which may help to understand the mechanism of promoting SCI recovery.
Collapse
Affiliation(s)
- Shiyuan Yin
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Feiyun Xia
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Wenjun Zou
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Fengxian Jiang
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Kelv Shen
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Baihan Sun
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Zhengfeng Lu
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China.
| |
Collapse
|
5
|
Xing X, Zhang S. Neuroprotective Role of AQP4 Knockdown in Astrocytes After Oxygen-Glucose Deprivation. Brain Behav 2024; 14:e70107. [PMID: 39444081 PMCID: PMC11499208 DOI: 10.1002/brb3.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/05/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Aquaporin-4 (AQP4), predominantly expressed in astrocytes, has been implicated in the development of brain edema following ischemic events. However, its role in post-stroke neuroinflammation is not fully understood. METHODS Using a middle cerebral artery occlusion (MCAO) mouse model, we assessed AQP4's role in post-stroke inflammation. Brain tissue slices from male C57BL/6 mice were subjected to immunohistochemistry and western blot post-MCAO. Additionally, primary astrocytes were isolated for quantitative real-time PCR and immunofluorescence assays to evaluate the expression of inflammatory markers glial fibrillary acidic protein (GFAP) and AQP4. AQP4 modulation was achieved using viral knockdown and overexpression methods. Neuronal damage was assessed using flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) tests in co-culture studies. RESULTS MCAO mice exhibited a significant upregulation in GFAP. This reactive astrogliosis corresponded with an elevation in inflammatory markers. AQP4 expression responded to this inflammatory trend, peaking at 6 h after OGD and returning to baseline levels at 24 and 48 h. Co-culture experiments revealed that AQP4(+) astrocytes exacerbated injury in OGD-treated neurons, as evidenced by increased TUNEL positivity and apoptotic events. Conversely, AQP4(-) astrocytes appeared to have a protective effect. Knockdown of AQP4 resulted in reduced post-OGD inflammatory response, whereas AQP4 overexpression intensified the injury to neurons post-OGD. In vivo experiments also confirmed that AQP4 inhibitor TGN-020 reduced and overexpression of AQP4 increased behavioral abnormalities and brain infarcts. CONCLUSION Our findings underscore AQP4's pivotal role in modulating post-stroke neuroinflammation. Targeting AQP4 may present a novel therapeutic avenue for mitigating ischemia-induced neuronal damage.
Collapse
Affiliation(s)
- Xin Xing
- Department of NeurologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Shuyan Zhang
- Department of NeurologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
6
|
Zhang L, Zhao G, Luo Z, Yu Z, Liu G, Su G, Tang X, Yuan Z, Huang C, Sun HS, Feng ZP, Huang Z. AD16 attenuates neuroinflammation induced by cerebral ischemia through down-regulating astrocytes A1 polarization. Biomed Pharmacother 2024; 178:117209. [PMID: 39094544 DOI: 10.1016/j.biopha.2024.117209] [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: 02/16/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
A1 polarization of astrocytes mediated prolonged inflammation contributing to brain injury in ischemic stroke. We have previously shown that AD16 protects against neonatal hypoxic-ischemic brain damage in vivo and oxygen-glucose deprivation in vitro. More recently, AD16 has demonstrated safety, tolerability, and favorable pharmacokinetics in a randomized controlled phase I trial. In this study, we utilized a rat model of transient middle cerebral artery occlusion (tMCAO) to explore whether the anti-inflammatory compound AD16 protects against ischemic brain injury by regulating A1 polarization and its underlying mechanisms. Our results showed that AD16 treatment significantly reduced the brain infarcted volume and improved neurological function in tMCAO rats. GO analysis results show that differential genes among the Sham, tMCAO and AD16 treatment groups are involved in the regulation of cytokine and inflammatory response. KEGG enrichment pathways analysis mainly enriched in cytokine-cytokine receptor interaction, viral protein interaction with cytokine-cytokine receptor, TNF, chemokine, NF-κB and IL-17 signaling pathway. Furthermore, AD16 treatment decreased the permeability of the blood-brain barrier and suppressed neuroinflammation. AD16 treatment also significantly reduced the polarization of A1 and inhibited NF-κB and JAK2/STAT3 signaling pathways. This study demonstrates that AD16 protects against brain injury in ischemic stroke by reducing A1 polarization to suppress neuroinflammation through downregulating NF-κB and JAK2/STAT3 signaling. Our findings uncover a potential molecular mechanism for AD16 and suggest that AD16 holds promising therapeutic potential against cerebral ischemia.
Collapse
Affiliation(s)
- Limei Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China; School of Basic Medical Sciences, China Medical University, Shengyang 110122, China
| | - Guojian Zhao
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Zhengwei Luo
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zining Yu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Gaigai Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Guangjun Su
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Xiaolu Tang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Zhidong Yuan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Cheng Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Zhihua Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Ganzhou Key Laboratory of Neuroinflammation Research, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China; School of Basic Medical Sciences, China Medical University, Shengyang 110122, China.
| |
Collapse
|
7
|
Liu K, Yang Y, Wu Z, Sun C, Su Y, Huang N, Wu H, Yi C, Ye J, Xiao L, Niu J. Remyelination-oriented clemastine treatment attenuates neuropathies of optic nerve and retina in glaucoma. Glia 2024; 72:1555-1571. [PMID: 38829008 DOI: 10.1002/glia.24543] [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: 11/28/2023] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024]
Abstract
As one of the top causes of blindness worldwide, glaucoma leads to diverse optic neuropathies such as degeneration of retinal ganglion cells (RGCs). It is widely accepted that the level of intraocular pressure (IOP) is a major risk factor in human glaucoma, and reduction of IOP level is the principally most well-known method to prevent cell death of RGCs. However, clinical studies show that lowering IOP fails to prevent RGC degeneration in the progression of glaucoma. Thus, a comprehensive understanding of glaucoma pathological process is required for developing new therapeutic strategies. In this study, we provide functional and histological evidence showing that optic nerve defects occurred before retina damage in an ocular hypertension glaucoma mouse model, in which oligodendroglial lineage cells were responsible for the subsequent neuropathology. By treatment with clemastine, an Food and Drug Administration (FDA)-approved first-generation antihistamine medicine, we demonstrate that the optic nerve and retina damages were attenuated via promoting oligodendrocyte precursor cell (OPC) differentiation and enhancing remyelination. Taken together, our results reveal the timeline of the optic neuropathies in glaucoma and highlight the potential role of oligodendroglial lineage cells playing in its treatment. Clemastine may be used in future clinical applications for demyelination-associated glaucoma.
Collapse
Affiliation(s)
- Kun Liu
- Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing, China
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Yujian Yang
- Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing, China
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Zhonghao Wu
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Chunhui Sun
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yixun Su
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Nanxin Huang
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Haoqian Wu
- Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jian Ye
- Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Jianqin Niu
- Department of Histology and Embryology, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
Wang L, Gao F, Chen L, Sun W, Liu H, Yang W, Zhang X, Bai J, Wang R. Remote Ischemia Postconditioning Mitigates Hippocampal Neuron Impairment by Modulating Cav1.2-CaMKIIα-Aromatase Signaling After Global Cerebral Ischemia in Ovariectomized Rats. Mol Neurobiol 2024; 61:6511-6527. [PMID: 38321351 PMCID: PMC11339123 DOI: 10.1007/s12035-024-03930-1] [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: 10/11/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024]
Abstract
Brain-derived estrogen (BDE2) is gaining attention as an endogenous neurotransmitter. Recent research has revealed that selectively removing the aromatase gene, the pivotal enzyme responsible for BDE2 synthesis, in forebrain neurons or astrocytes can lead to synaptic loss and cognitive impairment. It is worth noting that remote ischemia post-conditioning (RIP), a non-invasive technique, has been shown to activate natural protective mechanisms against severe ischemic events. The aim of our study was to investigate whether RIP triggers aromatase-BDE2 signaling, shedding light on its neuroprotective mechanisms after global cerebral ischemia (GCI) in ovariectomized rats. Our findings are as follows: (1) RIP was effective in mitigating ischemic damage in hippocampal CA1 neurons and improved cognitive function after GCI. This was partially due to increased Aro-BDE2 signaling in CA1 neurons. (2) RIP intervention efficiently enhanced pro-survival kinase pathways, such as AKT, ERK1/2, CREB, and suppressed CaMKIIα signaling in CA1 astrocytes induced by GCI. Remarkably, inhibiting CaMKIIα activity led to elevated Aro-BDE2 levels and replicated the benefits of RIP. (3) We also identified the positive mediation of Cav1.2, an LVGCC calcium channel, on CaMKIIα-Aro/BDE2 pathway response to RIP intervention. (4) Significantly, either RIP or CaMKIIα inhibition was found to alleviate reactive astrogliosis, which was accompanied by increased pro-survival A2-astrocyte protein S100A10 and decreased pro-death A1-astrocyte marker C3 levels. In summary, our study provides compelling evidence that Aro-BDE2 signaling is a critical target for the reparative effects of RIP following ischemic insult. This effect may be mediated through the CaV1.2-CaMKIIα signaling pathway, in collaboration with astrocyte-neuron interactions, thereby maintaining calcium homeostasis in the neuronal microenvironment and reducing neuronal damage after ischemia.
Collapse
Affiliation(s)
- Lu Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Lingling Chen
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Wuxiang Sun
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Huiyu Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Wei Yang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Xin Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
- Dementia and Dyscognitive Key Lab., North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210, Hebei, China.
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, Hebei, China.
| |
Collapse
|
9
|
Sippola S, Todorović M, Peltola E. First-Principles Structure Search Study of 17-β-Estradiol Adsorption on Graphene. ACS OMEGA 2024; 9:34684-34691. [PMID: 39157074 PMCID: PMC11325392 DOI: 10.1021/acsomega.4c03485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024]
Abstract
17-Beta-estradiol (E2), a steroid hormone synthesized from cholesterol, has various impacts on health and the environment. Currently, the gold standard for its measurement in the body is a conventional blood test (mass spectrometry), but carbon-based electrochemical sensors have been proposed as an alternative due to their advantages, such as rapid analysis time and sensitivity. To improve the atomic-level understanding of the interactions at the substrate surface, we performed density functional theory (DFT) simulations to study the nature of the adsorption of E2 on pristine graphene. Bayesian Optimization Structure Search (BOSS) was employed to reduce human bias in the determination of the most favorable adsorption configurations. Two stable adsorption minimum configurations were found. Analysis of their electronic properties indicates that E2 physisorbs on graphene. Embarking upon complex carbonaceous materials, the importance of finding all possible minimum candidates with automated structure search tools is highlighted. Computational investigations facilitate tailoring substrate materials with outstanding performance and applications in neuroscientific research, fertility monitoring, and clinical trials. Combining them with experimental research carries significant potential to advance sensor design beyond the current state-of-the-art.
Collapse
Affiliation(s)
- Saara Sippola
- Department of Mechanical
and Materials Engineering, University of
Turku, Turku 20500, Finland
| | - Milica Todorović
- Department of Mechanical
and Materials Engineering, University of
Turku, Turku 20500, Finland
| | - Emilia Peltola
- Department of Mechanical
and Materials Engineering, University of
Turku, Turku 20500, Finland
| |
Collapse
|
10
|
Wang X, Feng S, Deng Q, Wu C, Duan R, Yang L. The role of estrogen in Alzheimer's disease pathogenesis and therapeutic potential in women. Mol Cell Biochem 2024:10.1007/s11010-024-05071-4. [PMID: 39088186 DOI: 10.1007/s11010-024-05071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Estrogens are pivotal regulators of brain function throughout the lifespan, exerting profound effects from early embryonic development to aging. Extensive experimental evidence underscores the multifaceted protective roles of estrogens on neurons and neurotransmitter systems, particularly in the context of Alzheimer's disease (AD) pathogenesis. Studies have consistently revealed a greater risk of AD development in women compared to men, with postmenopausal women exhibiting heightened susceptibility. This connection between sex factors and long-term estrogen deprivation highlights the significance of estrogen signaling in AD progression. Estrogen's influence extends to key processes implicated in AD, including amyloid precursor protein (APP) processing and neuronal health maintenance mediated by brain-derived neurotrophic factor (BDNF). Reduced BDNF expression, often observed in AD, underscores estrogen's role in preserving neuronal integrity. Notably, hormone replacement therapy (HRT) has emerged as a sex-specific and time-dependent strategy for primary cardiovascular disease (CVD) prevention, offering an excellent risk profile against aging-related disorders like AD. Evidence suggests that HRT may mitigate AD onset and progression in postmenopausal women, further emphasizing the importance of estrogen signaling in AD pathophysiology. This review comprehensively examines the physiological and pathological changes associated with estrogen in AD, elucidating the therapeutic potential of estrogen-based interventions such as HRT. By synthesizing current knowledge, it aims to provide insights into the intricate interplay between estrogen signaling and AD pathogenesis, thereby informing future research directions and therapeutic strategies for this debilitating neurodegenerative disorder.
Collapse
Affiliation(s)
- Xinyi Wang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Xu P, Liu Y, Wang J, Zhang A, Wang K, Wang Z, Fang Y, Wang X, Zhang J. Gender-specific prognosis models reveal differences in subarachnoid hemorrhage patients between sexes. CNS Neurosci Ther 2024; 30:e14894. [PMID: 39107957 PMCID: PMC11303446 DOI: 10.1111/cns.14894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) represents a severe stroke subtype. Our study aims to develop gender-specific prognostic prediction models derived from distinct prognostic factors observed among different-gender patients. METHODS Inclusion comprised SAH-diagnosed patients from January 2014 to March 2016 in our institution. Collected data encompassed patients' demographics, admission severity, treatments, imaging findings, and complications. Three-month post-discharge prognoses were obtained via follow-ups. Analyses assessed gender-based differences in patient information. Key factors underwent subgroup analysis, followed by univariate and multivariate analyses to identify gender-specific prognostic factors and establish/validate gender-specific prognostic models. RESULTS A total of 929 patients, with a median age of 57 (16) years, were analyzed; 372 (40%) were male, and 557 (60%) were female. Differences in age, smoking history, hypertension, aneurysm presence, and treatment interventions existed between genders (p < 0.01), yet no disparity in prognosis was noted. Subgroup analysis explored hypertension history, aneurysm presence, and treatment impact, revealing gender-specific variations in these factors' influence on the disease. Screening identified independent prognostic factors: age, SEBES score, admission GCS score, and complications for males; and age, admission GCS score, intraventricular hemorrhage, treatment interventions, symptomatic vasospasm, hydrocephalus, delayed cerebral ischemia, and seizures for females. Evaluation and validation of gender-specific models yielded an AUC of 0.916 (95% CI: 0.878-0.954) for males and 0.914 (95% CI: 0.885-0.944) for females in the ROC curve. Gender-specific prognostic models didn't significantly differ from the overall population-based model (model 3) but exhibited robust discriminative ability and clinical utility. CONCLUSION Variations in baseline and treatment-related factors among genders contribute partly to gender-based prognosis differences. Independent prognostic factors vary by gender. Gender-specific prognostic models exhibit favorable prognostic performance.
Collapse
Affiliation(s)
- Penglei Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yuchun Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
| | - Junjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of MedicineZhejiang University School of MedicineYiwuChina
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Kaikai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Zefeng Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
- Brain Research InstituteZhejiang UniversityZhejiangChina
- MOE Frontier Science Center for Brain Science & Brain‐Machine IntegrationZhejiang UniversityZhejiangChina
| |
Collapse
|
12
|
Liu XH, Zhang LY, Liu XY, Zhang JG, Hu YY, Zhao CG, Xian XH, Li WB, Zhang M. Transformation of A1/A2 Astrocytes Participates in Brain Ischemic Tolerance Induced by Cerebral Ischemic Preconditioning via Inhibiting NDRG2. Neurochem Res 2024; 49:1665-1676. [PMID: 38411782 DOI: 10.1007/s11064-024-04134-8] [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: 08/06/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Cerebral ischemic preconditioning (CIP) has been shown to improve brain ischemic tolerance against subsequent lethal ischemia. Reactive astrocytes play important roles in cerebral ischemia-reperfusion. Recent studies have shown that reactive astrocytes can be polarized into neurotoxic A1 phenotype (C3d) and neuroprotective A2 phenotype (S100A10). However, their role in CIP remains unclear. Here, we focused on the role of N-myc downstream-regulated gene 2 (NDRG2) in regulating the transformation of A1/A2 astrocytes and promoting to brain ischemic tolerance induced by CIP. A Sprague Dawley rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) was used. Rats were divided into the following six groups: (1) sham group; (2) CIP group: left middle cerebral artery was blocked for 10 min; (3) MCAO/R group: left middle cerebral artery was blocked for 90 min; (4) CIP + MCAO/R group: CIP was performed 72 h before MCAO/R; (5) AAV-NDRG2 + CIP + MCAO/R group: adeno-associated virus (AAV) carrying NDRG2 was administered 14 days before CIP + MCAO/R; (6) AAV-Ctrl + CIP + MCAO/R group: empty control group. The rats were subjected to neurological evaluation 24 h after the above treatments, and then were sacrificed for 2, 3, 5-triphenyltetraolium chloride staining, thionin staining, immunofluorescence and western blot analysis. In CIP + MCAO/R group, the neurological deficit scores decreased, infarct volume reduced, and neuronal density increased compared with MCAO/R group. Notably, CIP significantly increased S100A10 expression and the number of S100A10+/GFAP+ cells, and also increased NDRG2 expression. MCAO/R significantly decreased S100A10 expression and the number of S100A10+/GFAP+ cells yet increased C3d expression and the number of C3d+/GFAP+ cells and NDRG2 expression, and these trends were reversed by CIP + MCAO/R. Furthermore, over-expression of NDRG2 before CIP + MCAO/R, the C3d expression and the number of C3d+/GFAP+ cells increased, while S100A10 expression and the number of S100A10+/GFAP+ cells decreased. Meanwhile, over-expression of NDRG2 blocked the CIP-induced brain ischemic tolerance. Taken together, these results suggest that CIP exerts neuroprotective effects against ischemic injury by suppressing A1 astrocyte polarization and promoting A2 astrocyte polarization via inhibiting NDRG2 expression.
Collapse
Affiliation(s)
- Xiao-Hua Liu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Department of Physiology, Shijiazhuang Medical College, Shijiazhuang, 050000, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China.
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Chen-Guang Zhao
- Department of foreign language, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
13
|
Santiago-Balmaseda A, Aguirre-Orozco A, Valenzuela-Arzeta IE, Villegas-Rojas MM, Pérez-Segura I, Jiménez-Barrios N, Hurtado-Robles E, Rodríguez-Hernández LD, Rivera-German ER, Guerra-Crespo M, Martinez-Fong D, Ledesma-Alonso C, Diaz-Cintra S, Soto-Rojas LO. Neurodegenerative Diseases: Unraveling the Heterogeneity of Astrocytes. Cells 2024; 13:921. [PMID: 38891053 PMCID: PMC11172252 DOI: 10.3390/cells13110921] [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: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The astrocyte population, around 50% of human brain cells, plays a crucial role in maintaining the overall health and functionality of the central nervous system (CNS). Astrocytes are vital in orchestrating neuronal development by releasing synaptogenic molecules and eliminating excessive synapses. They also modulate neuronal excitability and contribute to CNS homeostasis, promoting neuronal survival by clearance of neurotransmitters, transporting metabolites, and secreting trophic factors. Astrocytes are highly heterogeneous and respond to CNS injuries and diseases through a process known as reactive astrogliosis, which can contribute to both inflammation and its resolution. Recent evidence has revealed remarkable alterations in astrocyte transcriptomes in response to several diseases, identifying at least two distinct phenotypes called A1 or neurotoxic and A2 or neuroprotective astrocytes. However, due to the vast heterogeneity of these cells, it is limited to classify them into only two phenotypes. This review explores the various physiological and pathophysiological roles, potential markers, and pathways that might be activated in different astrocytic phenotypes. Furthermore, we discuss the astrocyte heterogeneity in the main neurodegenerative diseases and identify potential therapeutic strategies. Understanding the underlying mechanisms in the differentiation and imbalance of the astrocytic population will allow the identification of specific biomarkers and timely therapeutic approaches in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Alberto Santiago-Balmaseda
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Annai Aguirre-Orozco
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Irais E. Valenzuela-Arzeta
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Marcos M. Villegas-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico
| | - Isaac Pérez-Segura
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Natalie Jiménez-Barrios
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Ernesto Hurtado-Robles
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Luis Daniel Rodríguez-Hernández
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Erick R. Rivera-German
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Magdalena Guerra-Crespo
- Laboratorio de Medicina Regenerativa, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico;
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| |
Collapse
|
14
|
Wang M, Hu S, Fu X, Zhou H, Yang S, Yang C. Neurosteroids: A potential target for neuropsychiatric disorders. J Steroid Biochem Mol Biol 2024; 239:106485. [PMID: 38369032 DOI: 10.1016/j.jsbmb.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Neurosteroids are steroids produced by endocrine glands and subsequently entering the brain, and also include steroids synthesis in the brain. It has been widely known that neurosteroids influence many neurological functions, including neuronal signaling, synaptic adaptations, and neuroprotective effects. In addition, abnormality in the synthesis and function of neurosteroids has been closely linked to neuropsychiatric disorders, such as Alzheimer's disease (AD), schizophrenia (SZ), and epilepsy. Given their important role in brain pathophysiology and disorders, neurosteroids offer potential therapeutic targets for a variety of neuropsychiatric diseases, and that therapeutic strategies targeting neurosteroids probably exert beneficial effects. We therefore summarized the role of neurosteroids in brain physiology and neuropsychiatric disorders, and introduced the recent findings of synthetic neurosteroid analogues for potential treatment of neuropsychiatric disorders, thereby providing insights for further research in the future.
Collapse
Affiliation(s)
- Mengyu Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinghuo Fu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Huixuan Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
15
|
Stennette KA, Godwin JR. Estrogenic influences on agonistic behavior in teleost fishes. Horm Behav 2024; 161:105519. [PMID: 38452611 DOI: 10.1016/j.yhbeh.2024.105519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024]
Abstract
Teleost fishes show an extraordinary diversity of sexual patterns, social structures, and sociosexual behaviors. Sex steroid hormones are key modulators of social behaviors in teleosts as in other vertebrates and act on sex steroid receptor-containing brain nuclei that form the evolutionarily conserved vertebrate social behavior network (SBN). Fishes also display important differences relative to tetrapod vertebrates that make them particularly well-suited to study the physiological mechanisms modulating social behavior. Specifically, fishes exhibit high levels of brain aromatization and have what has been proposed to be a lifelong, steroid hormone dependent plasticity in the neural substrates mediating sociosexual behavior. In this review, we examine how estrogenic signaling modulates sociosexual behaviors in teleosts with a particular focus on agonistic behavior. Estrogens have been shown to mediate agonistic behaviors in a broad range of fishes, from sexually monomorphic gonochoristic species to highly dimorphic sex changers with alternate reproductive phenotypes. These similarities across such diverse taxa contribute to a growing body of evidence that estrogens play a crucial role in the modulation of aggression in vertebrates. As analytical techniques and genomic tools rapidly advance, methods such as LC-MS/MS, snRNAseq, and CRISPR-based mutagenesis show great promise to further elucidate the mechanistic basis of estrogenic effects on social behavior in the diverse teleost lineage.
Collapse
Affiliation(s)
- Katherine A Stennette
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - John R Godwin
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
16
|
Li B, Yu W, Verkhratsky A. Trace metals and astrocytes physiology and pathophysiology. Cell Calcium 2024; 118:102843. [PMID: 38199057 DOI: 10.1016/j.ceca.2024.102843] [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: 12/18/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Several trace metals, including iron, copper, manganese and zinc are essential for normal function of the nervous system. Both deficiency and excessive accumulation of these metals trigger neuropathological developments. The central nervous system (CNS) is in possession of dedicated homeostatic system that removes, accumulates, stores and releases these metals to fulfil nervous tissue demand. This system is mainly associated with astrocytes that act as dynamic reservoirs for trace metals, these being a part of a global system of CNS ionostasis. Here we overview physiological and pathophysiological aspects of astrocyte-cantered trace metals regulation.
Collapse
Affiliation(s)
- Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China; China Medical University Centre of Forensic Investigation, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China; China Medical University Centre of Forensic Investigation, China
| | - Alexei Verkhratsky
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, Ikerbasque, Bilbao 48011, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius LT-01102, Lithuania.
| |
Collapse
|
17
|
Supakul S, Oyama C, Hatakeyama Y, Maeda S, Okano H. Estradiol enhanced neuronal plasticity and ameliorated astrogliosis in human iPSC-derived neural models. Regen Ther 2024; 25:250-263. [PMID: 38293585 PMCID: PMC10826128 DOI: 10.1016/j.reth.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/30/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction 17β-Estradiol (E2) is a sex hormone that has been previously demonstrated to have neurotherapeutic effects on animal models of Alzheimer's disease (AD). However, clinical trials on E2 replacement therapy for preventing AD onset yielded inconsistent results. Therefore, it is imperative to clarify the therapeutic effects of E2 on human cells. In this study, we utilized induced pluripotent stem cells (iPSCs) derived from multiple AD donors to explore the therapeutic effects of E2 on the in vitro model of human cells. Methods We conducted a systematic review and meta-analysis using a random-effects model of the previously reported AD clinical trials to summarize the effects of E2 replacement therapy on AD prevention. Subsequently, we induced iPSCs from the donors of the healthy control (1210B2 line (female) and 201B7 line (female)), the familial AD (APP V717L line (female) and APP KM670/671NL line (female)), and the sporadic AD (UCSD-SAD3.7 line (APOE ε3/ε3) (male), UCSD-SAD7D line (APOE ε3/ε4) (male), and TMGH-1 line (APOE ε3/ε3) (female)), then differentiated to neurons. In addition to the mono-culture model of the neurons, we also examined the effects of E2 on the co-culture model of neurons and astrocytes. Results The meta-analysis of the clinical trials concluded that E2 replacement therapy reduced the risk of AD onset (OR, 0.69; 95 % confidence interval [CI], 0.53-0.91; I2 = 82 %). Neural models from the iPSCs of AD donors showed an increase in secreted amyloid-beta (Aβ) levels in the mono-culture model and an astrogliosis-like phenotype in the co-culture model. E2 treatment to the neuronal models derived from the iPSCs enhanced neuronal activity and increased neurite complexity. Furthermore, E2 treatment of the co-culture model ameliorated the astrogliosis-like phenotype. However, in contrast to the previous reports using mouse models, E2 treatment did not change AD pathogenesis, including Aβ secretion and phosphorylated tau (pTau) accumulation. Conclusion E2 treatment of the human cellular model did not impact Aβ secretion and pTau accumulation, but promoted neuronal plasticity and alleviated the astrogliosis-like phenotype. The limited effects of E2 may give a clue for the mixed results of E2 clinical trials.
Collapse
Affiliation(s)
- Sopak Supakul
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Chisato Oyama
- Department of Electrical Engineering and Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuki Hatakeyama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Sumihiro Maeda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Tan XX, Dai HY, Yao J, Wang JJ, Dai YC, Zhang TH, Qiu LL, Sun J. Hippocampal estrogens rescued the decline of synaptic plasticity after surgery and anesthesia by inhibiting microglia overactivation. Behav Brain Res 2024; 459:114794. [PMID: 38056710 DOI: 10.1016/j.bbr.2023.114794] [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: 07/22/2023] [Revised: 10/30/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Elderly patients experience postoperative cognitive impairment frequently; therefore, effective interventions are urgently needed. Central nervous inflammation characterized by microglia may promote the progression of POCD by reducing synaptic plasticity. Notably, clinical studies revealed that the incidence of female patients was significantly lower than that of male patients. Besides, the brain estrogens have an anti-inflammatory effect and regulate the microglia at the same times. This study aimed to determine whether suppressing microglia overactivation by hippocampal estrogens can rescue the decrease of synaptic plasticity after surgery and anesthesia. METHODS Exploratory laparotomy was used to establish the POCD model in 15-month-old male or female C57BL/6 J mice and animal behavioral tests were performed to test hippocampal-dependent memory capacity. Western blot and immunofluorescence were used to detect the microglial activation and plasticity related protein expressions. Elisa was used to detect the content of estrogens in the hippocampus. Estrogens and estrogen receptor inhibitor were used to replenish the estrogens in the brain and inhibit the effect of estrogens. RESULTS Surgery and anesthesia did not cause POCD in female mice (P > 0.05), while the cognitive function decreased significantly after estrogen receptor inhibitor was given(P < 0.05). Male mice experienced cognitive dysfunction after surgery and anesthesia, and their cognitive function improved after estrogens supplementation (P < 0.05). Given estrogens and estrogen receptor inhibitors at the same time, the cognitive function of male mice could not be saved (P < 0.05). By correlation analysis, there was a negative correlation between the content of hippocampal estrogens and microglia (P < 0.05). The number or degree of activation of microglia affected the synaptic plasticity, which ultimately regulated the cognitive function of mice. CONCLUSION Hippocampal estrogens rescued the decline of synaptic plasticity after surgery and anesthesia by inhibiting microglia overactivation.
Collapse
Affiliation(s)
- Xiao-Xiang Tan
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Hong-Yu Dai
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Juan Yao
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Jia-Jia Wang
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Yu-Chen Dai
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Tian-Hao Zhang
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Li-Li Qiu
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China.
| | - Jie Sun
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
19
|
Czyżewski W, Mazurek M, Sakwa L, Szymoniuk M, Pham J, Pasierb B, Litak J, Czyżewska E, Turek M, Piotrowski B, Torres K, Rola R. Astroglial Cells: Emerging Therapeutic Targets in the Management of Traumatic Brain Injury. Cells 2024; 13:148. [PMID: 38247839 PMCID: PMC10813911 DOI: 10.3390/cells13020148] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic Brain Injury (TBI) represents a significant health concern, necessitating advanced therapeutic interventions. This detailed review explores the critical roles of astrocytes, key cellular constituents of the central nervous system (CNS), in both the pathophysiology and possible rehabilitation of TBI. Following injury, astrocytes exhibit reactive transformations, differentiating into pro-inflammatory (A1) and neuroprotective (A2) phenotypes. This paper elucidates the interactions of astrocytes with neurons, their role in neuroinflammation, and the potential for their therapeutic exploitation. Emphasized strategies encompass the utilization of endocannabinoid and calcium signaling pathways, hormone-based treatments like 17β-estradiol, biological therapies employing anti-HBGB1 monoclonal antibodies, gene therapy targeting Connexin 43, and the innovative technique of astrocyte transplantation as a means to repair damaged neural tissues.
Collapse
Affiliation(s)
- Wojciech Czyżewski
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Leon Sakwa
- Student Scientific Society, Kazimierz Pulaski University of Radom, 26-600 Radom, Poland;
| | - Michał Szymoniuk
- Student Scientific Association, Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Jennifer Pham
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Barbara Pasierb
- Department of Dermatology, Radom Specialist Hospital, 26-600 Radom, Poland;
| | - Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Czyżewska
- Department of Otolaryngology, Mazovian Specialist Hospital, 26-617 Radom, Poland;
| | - Michał Turek
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Bartłomiej Piotrowski
- Institute of Automatic Control and Robotics, Warsaw University of Technology, 00-661 Warsaw, Poland;
| | - Kamil Torres
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Radosław Rola
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| |
Collapse
|
20
|
Li L, Lin Z, Yuan J, Li P, Wang Q, Cho N, Wang Y, Lin Z. The neuroprotective mechanisms of naringenin: Inhibition of apoptosis through the PI3K/AKT pathway after hypoxic-ischemic brain damage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116941. [PMID: 37480970 DOI: 10.1016/j.jep.2023.116941] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naringenin (NGN) is a widely distributed flavonoid with potent antioxidant and neuroprotective properties. Neuroprotective agents play a crucial role in the treatment of hypoxic-ischemic encephalopathy (HIE). It has shown potential therapeutic effects for neurological disorders. However, its efficacy on HIE is yet to be investigated. AIM OF THE STUDY This study aims to investigate the potential neuroprotective effect of naringenin and its underlying molecular mechanisms in reducing oxidative stress, apoptosis, and improving brain outcomes following HIE. Additionally, the study aims to identify the potential targets, mechanisms, and functions of naringenin using network pharmacology analysis. MATERIALS AND METHODS Neonatal mice were exposed to the hypoxic-ischemic brain damage (HIBD) model to determine brain water content, and brain tissue was subjected to hematoxylin and eosin (HE), immunohistochemistry (IHC), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and Nissl staining to investigate its neuroprotective effects. Furthermore, the neonatal mouse primary neuron oxygen-glucose deprivation (OGD) model to measure reactive oxygen species (ROS) production in vitro. The protein levels were characterized by Western Blot, and mRNA levels were evaluated by a real-time quantitative PCR detecting system (qPCR). Transmission electron microscopy (TEM) and mitochondrial fluorescent staining were used to observe mitochondrial morphology. Neuronal nuclei (NeuN) and microtubule-associated protein 2 (MAP2) were detected by Immunofluorescence (IF). Finally, network pharmacology was employed to determine the common target of naringenin and HIE. The core genes were obtained via protein-protein interaction networks (PPI) analysis and molecular docking was examined, and the mechanism of action was explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Additionally, small interfering RNA (siRNA) was constructed for verification. RESULTS Naringenin has a neuroprotective effect in HIBD by modulating Vegfa expression and activating the PI3K/AKT pathway to inhibit apoptosis. Furthermore, molecular docking results suggest that Vegfa is a potential binding target of naringenin, and silencing Vegfa partially reverses the pharmacological effects of NGN. CONCLUSION Our findings suggest that naringenin demonstrates potential clinical application for treating HIE as a novel neuroprotective agent.
Collapse
Affiliation(s)
- Luyao Li
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Junhui Yuan
- Wenling Maternal and Child Health Care Hospital, Xiabao Road, Chengdong Street of Wenling City, Zhejiang Province, 317500, China
| | - Pingping Li
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Qi Wang
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju, South Korea.
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Zhenlang Lin
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
21
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Song T, Zhang Y, Zhu L, Zhang Y, Song J. The role of JAK/STAT signaling pathway in cerebral ischemia-reperfusion injury and the therapeutic effect of traditional Chinese medicine: A narrative review. Medicine (Baltimore) 2023; 102:e35890. [PMID: 37986307 PMCID: PMC10659620 DOI: 10.1097/md.0000000000035890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023] Open
Abstract
Cerebral ischemia is a cerebrovascular disease with symptoms caused by insufficient blood or oxygen supply to the brain. When blood supplied is restored after cerebral ischemia, secondary brain injury may occur, which is called cerebral ischemia-reperfusion injury (CIRI). In this process, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway plays an important role. It mediates neuroinflammation and participates in the regulation of physiological activities, such as cell proliferation, differentiation, and apoptosis. After CIRI, M1 microglia is activated and recruited by the damaged tissue. The inflammatory factors are produced by M1 microglia through the JAK/STAT pathway, eventually leading to cell apoptosis. Meanwhile, the JAK2/STAT3 signaling pathway and the expression of lipocalin-2 and caspase-3 could increase. In the pathway, phosphorylated JAK2 and phosphorylated STAT3 function of 2 ways. They not only promote the proliferation of neurons, but also affect the differentiation direction of neural stem cells by further acting on the Notch signaling pathway. Recently, traditional Chinese medicine (TCM) is a key player in CIRI, through JAK2, STAT3, STAT1 and their phosphorylation. Therefore, the review focuses on the JAK/STAT signaling pathway and its relationship with CIRI as well as the influence of the TCM on this pathway. It is aimed at providing the basis for future clinical research on the molecular mechanism of TCM in the treatment of CIRI.
Collapse
Affiliation(s)
- Tianzhi Song
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yishu Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liangrong Zhu
- Wenling Hospital of Traditional Chinese Medicine, Taizhou, China
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingmei Song
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Machado MMF, Ático EM, Banin RM, Hirata BKS, Kempe PRG, Pedroso AP, Thomaz FM, Oyama LM, Ribeiro EB, Bueno AA, Cerutti SM, Telles MM. Ginkgo biloba extract modulates astrocytic and microglial recruitment in the hippocampus and hypothalamus of menopause-induced ovariectomized rats. Brain Res 2023; 1822:148659. [PMID: 39492489 DOI: 10.1016/j.brainres.2023.148659] [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: 08/01/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND Changes in steroid hormone levels associated with menopause are known to affect body composition, with increased accumulation of visceral fat and impaired actions of appetite-regulating neuropeptides. Anti-obesogenic, antioxidant, anti-inflammatory and neuromodulatory properties have been attributed to Ginkgo biloba extract (GbE) oral supplementation. HYPOTHESIS/PURPOSE We investigated in menopause-induced ovariectomized rats the effects of GbE oral supplementation on microglial reactivity and astrocyte recruitment in hippocampal and hypothalamic subregions involved in the regulation of feeding behavior and energy homeostasis. STUDY DESIGN/METHODS Ovariectomy (Ovx) or false-Ovx (Sham) surgery were performed in 2-month-old female Wistar rats. Sixty days after surgery, Ovx rats were gavaged daily for 14 days with either saline (Ovx + Veh) or GbE 500 mg/Kg (Ovx + GbE). Rats were subsequently sacrificed, brains harvested and subjected to immunohistochemistry and immunofluorescence analyses. RESULTS Ovx increased microglial reactivity in CA1, CA3 and dentate gyrus (DG) in the dorsal hippocampal formation (dHF), as well as in DG in the ventral hippocampal formation (vHF). Additionally, Ovx reduced astrocyte count in dHF CA3. The disturbances found in Ovx + Veh versus Sham were not found in Ovx + GbE versus Sham. Furthermore, higher astrocyte counts in DG of both dHF and vHF were found in Ovx + GbE as compared to Ovx + Veh. In the hypothalamus, Ovx + Veh showed reduced microglial reactivity in the arcuate (ARC) and ventromedial (VMH) nuclei as compared to Ovx + GbE. Ovx + GbE rats presented higher astrocyte counts in ARC compared to Sham rats. CONCLUSION Our results show for the first time in a rodent model of menopause that GbE supplementation modulates astrocyte and microglial recruitment and reactivity in hippocampal and hypothalamic subregions involved in feeding behavior and energy homeostasis. Future research employing other experimental models may further elucidate whether GbE supplementation possesses therapeutic properties upon glial cell reactivity to potentially alleviate changes in energy homeostasis associated with menopause.
Collapse
Affiliation(s)
- Meira M F Machado
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Esther M Ático
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Renata M Banin
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruna K S Hirata
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Paula R G Kempe
- Laboratory of Nerve Regeneration, Universidade de Campinas, Campinas, Brazil
| | - Amanda P Pedroso
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Thomaz
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Lila M Oyama
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Eliane B Ribeiro
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester WR2 6AJ, United Kingdom.
| | - Suzete M Cerutti
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Mônica M Telles
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil; Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Wang Y, Liu W, Geng P, Du W, Guo C, Wang Q, Zheng GQ, Jin X. Role of Crosstalk between Glial Cells and Immune Cells in Blood-Brain Barrier Damage and Protection after Acute Ischemic Stroke. Aging Dis 2023; 15:2507-2525. [PMID: 37962453 PMCID: PMC11567273 DOI: 10.14336/ad.2023.1010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Blood-brain barrier (BBB) damage is the main pathological basis for acute ischemic stroke (AIS)-induced cerebral vasogenic edema and hemorrhagic transformation (HT). Glial cells, including microglia, astrocytes, and oligodendrocyte precursor cells (OPCs)/oligodendrocytes (OLs) play critical roles in BBB damage and protection. Recent evidence indicates that immune cells also have an important role in BBB damage, vasogenic edema and HT. Therefore, regulating the crosstalk between glial cells and immune cells would hold the promise to alleviate AIS-induced BBB damage. In this review, we first introduce the roles of glia cells, pericytes, and crosstalk between glial cells in the damage and protection of BBB after AIS, emphasizing the polarization, inflammatory response and crosstalk between microglia, astrocytes, and other glia cells. We then describe the role of glial cell-derived exosomes in the damage and protection of BBB after AIS. Next, we specifically discuss the crosstalk between glial cells and immune cells after AIS. Finally, we propose that glial cells could be a potential target for alleviating BBB damage after AIS and we discuss some molecular targets and potential strategies to alleviate BBB damage by regulating glial cells after AIS.
Collapse
Affiliation(s)
- Yihui Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wencao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Guo-qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
25
|
Fiore NT, Keating BA, Chen Y, Williams SI, Moalem-Taylor G. Differential Effects of Regulatory T Cells in the Meninges and Spinal Cord of Male and Female Mice with Neuropathic Pain. Cells 2023; 12:2317. [PMID: 37759539 PMCID: PMC10527659 DOI: 10.3390/cells12182317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Immune cells play a critical role in promoting neuroinflammation and the development of neuropathic pain. However, some subsets of immune cells are essential for pain resolution. Among them are regulatory T cells (Tregs), a specialised subpopulation of T cells that limit excessive immune responses and preserve immune homeostasis. In this study, we utilised intrathecal adoptive transfer of activated Tregs in male and female mice after peripheral nerve injury to investigate Treg migration and whether Treg-mediated suppression of pain behaviours is associated with changes in peripheral immune cell populations in lymphoid and meningeal tissues and spinal microglial and astrocyte reactivity and phenotypes. Treatment with Tregs suppressed mechanical pain hypersensitivity and improved changes in exploratory behaviours after chronic constriction injury (CCI) of the sciatic nerve in both male and female mice. The injected Treg cells were detected in the choroid plexus and the pia mater and in peripheral lymphoid organs in both male and female recipient mice. Nonetheless, Treg treatment resulted in differential changes in meningeal and lymph node immune cell profiles in male and female mice. Moreover, in male mice, adoptive transfer of Tregs ameliorated the CCI-induced increase in microglia reactivity and inflammatory phenotypic shift, increasing M2-like phenotypic markers and attenuating astrocyte reactivity and neurotoxic astrocytes. Contrastingly, in CCI female mice, Treg injection increased astrocyte reactivity and neuroprotective astrocytes. These findings show that the adoptive transfer of Tregs modulates meningeal and peripheral immunity, as well as spinal glial populations, and alleviates neuropathic pain, potentially through different mechanisms in males and females.
Collapse
Affiliation(s)
| | | | | | | | - Gila Moalem-Taylor
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (N.T.F.); (B.A.K.); (Y.C.); (S.I.W.)
| |
Collapse
|
26
|
Hu J, Huang Y, Gao F, Sun W, Liu H, Ma H, Yuan T, Liu Z, Tang L, Ma Y, Zhang X, Bai J, Wang R. Brain-derived estrogen: a critical player in maintaining cognitive health of aged female rats, possibly involving GPR30. Neurobiol Aging 2023; 129:15-27. [PMID: 37257405 DOI: 10.1016/j.neurobiolaging.2023.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Brain-derived estrogen is an endogenous neuroprotective agent, whether and how might this protective function with aging, especially postmenopausal drops in circulating estrogen, remain unclear. We herein subjected 6, 14, and 18 Mon female rats to mimic natural aging, and found that estrogen synthesis is more active in the healthy aged brain, as evidenced by the highest levels of mRNA and protein expression of aromatase, the key enzyme of E2 biosynthesis, among the three groups. Aromatase knockout in forebrain neurons (FBN-Aro-/-) impaired hippocampal and cortical neurons, and cognitive function in 18 Mon rats, compared to wild-type controls. Furthermore, estrogen nuclear receptors (ERα/β) displayed opposite changes, with a significant ERα decrease and ERβ increase, while membrane receptor GPR30 expressed stably in hippocampus during aging. Intriguingly, GPR30, but not ERα and ERβ, was decreased by FBN-Aro-/-. The results indicate that GPR30 is more sensitive to brain local E2 synthesis. Our findings provide evidence of a critical role for brain-derived estrogen in maintaining healthy brain function in older individuals, possibly involving GPR30.
Collapse
Affiliation(s)
- Jiewei Hu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Yuanyuan Huang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Fujia Gao
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Wuxiang Sun
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Huiyu Liu
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Haoran Ma
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Tao Yuan
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Zixuan Liu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Lei Tang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Yuxuan Ma
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Xin Zhang
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Jing Bai
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| | - Ruimin Wang
- Neurobiology Institute, Key Laboratory of Dementia and Cognitive Dysfunction, School of Public Health of North China University of Science and Technology, Tangshan, Hebei, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, China.
| |
Collapse
|
27
|
Ugidos IF, González-Rodríguez P, Santos-Galdiano M, Font-Belmonte E, Anuncibay-Soto B, Pérez-Rodríguez D, Gonzalo-Orden JM, Fernández-López A. Neuroprotective effects of meloxicam on transient brain ischemia in rats: the two faces of anti-inflammatory treatments. Neural Regen Res 2023; 18:1961-1967. [PMID: 36926720 PMCID: PMC10233777 DOI: 10.4103/1673-5374.367846] [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: 08/23/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 01/22/2023] Open
Abstract
The inflammatory response plays an important role in neuroprotection and regeneration after ischemic insult. The use of non-steroidal anti-inflammatory drugs has been a matter of debate as to whether they have beneficial or detrimental effects. In this context, the effects of the anti-inflammatory agent meloxicam have been scarcely documented after stroke, but its ability to inhibit both cyclooxygenase isoforms (1 and 2) could be a promising strategy to modulate post-ischemic inflammation. This study analyzed the effect of meloxicam in a transient focal cerebral ischemia model in rats, measuring its neuroprotective effect after 48 hours and 7 days of reperfusion and the effects of the treatment on the glial scar and regenerative events such as the generation of new progenitors in the subventricular zone and axonal sprouting at the edge of the damaged area. We show that meloxicam's neuroprotective effects remained after 7 days of reperfusion even if its administration was restricted to the two first days after ischemia. Moreover, meloxicam treatment modulated glial scar reactivity, which matched with an increase in axonal sprouting. However, this treatment decreased the formation of neuronal progenitor cells. This study discusses the dual role of anti-inflammatory treatments after stroke and encourages the careful analysis of both the neuroprotective and the regenerative effects in preclinical studies.
Collapse
Affiliation(s)
- Irene Fernández Ugidos
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| | - Paloma González-Rodríguez
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| | - María Santos-Galdiano
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
- Neural Therapies SL. Edif. Institutos de Investigación. Planta baja. Local B43. Campus de Vegazana s/n. León. Spain
| | - Enrique Font-Belmonte
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| | - Berta Anuncibay-Soto
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| | - Diego Pérez-Rodríguez
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| | - José Manuel Gonzalo-Orden
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
- Department of Medicina, Cirugía y Anatomía Veterinaria, University of León, León, Spain
| | - Arsenio Fernández-López
- Área de Biología Celular, Instituto de Biomedicina, Campus de Vegazana s/n, Universidad de León, León, Spain
| |
Collapse
|
28
|
Wang J, Deng X, Jiang J, Yao Z, Ju Y, Luo Y. Evaluation of electroacupuncture as a non-pharmacological therapy for astrocytic structural aberrations and behavioral deficits in a post-ischemic depression model in mice. Front Behav Neurosci 2023; 17:1239024. [PMID: 37700911 PMCID: PMC10493307 DOI: 10.3389/fnbeh.2023.1239024] [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: 06/27/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Background Ascending clinical evidence supports that electroacupuncture (EA) is effective in treating post-ischemic depression (PID), but little is known about how it works at the cellular level. Astrocytes are exquisitely sensitive to their extracellular environment, and under stressful conditions, they may experience aberrant structural remodeling that can potentially cause neuroplastic disturbances and contribute to subsequent changes in mood or behavior. Objectives This study aimed to investigate the effect of EA on behavioral deficits associated with PID in mice and verify the hypothesis that astrocytic morphology may be involved in this impact. Methods We established a PID animal model induced by transient bilateral common carotid artery occlusion (BCCAO, 20 min) and chronic restraint stress (CRS, 21 days). EA treatment (GV20 + ST36) was performed for 3 weeks, from Monday to Friday each week. Depressive- and anxiety-like behaviors and sociability were evaluated using SPT, FST, EPM, and SIT. Immunohistochemistry combined with Sholl and cell morphological analysis was utilized to assess the process morphology of GFAP+ astrocytes in mood-related regions. The potential relationship between morphological changes in astrocytes and behavioral output was detected by correlation analysis. Results Behavioral assays demonstrated that EA treatment induced an overall reduction in behavioral deficits, as measured by the behavioral Z-score. Sholl and morphological analyses revealed that EA prevented the decline in cell complexity of astrocytes in the prefrontal cortex (PFC) and the CA1 region of the hippocampus, where astrocytes displayed evident deramification and atrophy of the branches. Eventually, the correlation analysis showed there was a relationship between behavioral emotionality and morphological changes. Conclusion Our findings imply that EA prevents both behavioral deficits and structural abnormalities in astrocytes in the PID model. The strong correlation between behavioral Z-scores and the observed morphological changes confirms the notion that the weakening of astrocytic processes may play a crucial role in depressive symptoms, and astrocytes could be a potential target of EA in the treatment of PID.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengyu Yao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxin Ju
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
29
|
Huang B, Chen K, Li Y. Aerobic exercise, an effective prevention and treatment for mild cognitive impairment. Front Aging Neurosci 2023; 15:1194559. [PMID: 37614470 PMCID: PMC10442561 DOI: 10.3389/fnagi.2023.1194559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Abstract
Aerobic exercise has emerged as a promising intervention for mild cognitive impairment (MCI), a precursor to dementia. The therapeutic benefits of aerobic exercise are multifaceted, encompassing both clinical and molecular domains. Clinically, aerobic exercise has been shown to mitigate hypertension and type 2 diabetes mellitus, conditions that significantly elevate the risk of MCI. Moreover, it stimulates the release of nitric oxide, enhancing arterial elasticity and reducing blood pressure. At a molecular level, it is hypothesized that aerobic exercise modulates the activation of microglia and astrocytes, cells crucial to brain inflammation and neurogenesis, respectively. It has also been suggested that aerobic exercise promotes the release of exercise factors such as irisin, cathepsin B, CLU, and GPLD1, which could enhance synaptic plasticity and neuroprotection. Consequently, regular aerobic exercise could potentially prevent or reduce the likelihood of MCI development in elderly individuals. These molecular mechanisms, however, are hypotheses that require further validation. The mechanisms of action are intricate, and further research is needed to elucidate the precise molecular underpinnings and to develop targeted therapeutics for MCI.
Collapse
Affiliation(s)
- Baiqing Huang
- Sports Institute, Yunnan Minzu University, Kunming, China
| | - Kang Chen
- Tianjin Key Lab of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| | - Ying Li
- Sports Institute, Yunnan Minzu University, Kunming, China
| |
Collapse
|
30
|
Carvalho D, Diaz-Amarilla P, Dapueto R, Santi MD, Duarte P, Savio E, Engler H, Abin-Carriquiry JA, Arredondo F. Transcriptomic Analyses of Neurotoxic Astrocytes Derived from Adult Triple Transgenic Alzheimer's Disease Mice. J Mol Neurosci 2023; 73:487-515. [PMID: 37318736 DOI: 10.1007/s12031-023-02105-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/03/2023] [Indexed: 06/16/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease have been classically studied from a purely neuronocentric point of view. More recent evidences support the notion that other cell populations are involved in disease progression. In this sense, the possible pathogenic role of glial cells like astrocytes is increasingly being recognized. Once faced with tissue damage signals and other stimuli present in disease environments, astrocytes suffer many morphological and functional changes, a process referred as reactive astrogliosis. Studies from murine models and humans suggest that these complex and heterogeneous responses could manifest as disease-specific astrocyte phenotypes. Clear understanding of disease-associated astrocytes is a necessary step to fully disclose neurodegenerative processes, aiding in the design of new therapeutic and diagnostic strategies. In this work, we present the transcriptomics characterization of neurotoxic astrocytic cultures isolated from adult symptomatic animals of the triple transgenic mouse model of Alzheimer's disease (3xTg-AD). According to the observed profile, 3xTg-AD neurotoxic astrocytes show various reactivity features including alteration of the extracellular matrix and release of pro-inflammatory and proliferative factors that could result in harmful effects to neurons. Moreover, these alterations could be a consequence of stress responses at the endoplasmic reticulum and mitochondria as well as of concomitant metabolic adaptations. Present results support the hypothesis that adaptive changes of astrocytic function induced by a stressed microenvironment could later promote harmful astrocyte phenotypes and further accelerate or induce neurodegenerative processes.
Collapse
Affiliation(s)
- Diego Carvalho
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Pablo Diaz-Amarilla
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Rosina Dapueto
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - María Daniela Santi
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
- College of Dentistry, Bluestone Center for Clinical Research, New York University, New York, 10010, USA
| | - Pablo Duarte
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Eduardo Savio
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Henry Engler
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
- Facultad de Medicina, Universidad de la República, 1800, Montevideo, Uruguay
| | - Juan A Abin-Carriquiry
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay.
- Laboratorio de Biofármacos, Institut Pasteur de Montevideo, 11600, Montevideo, Uruguay.
| | - Florencia Arredondo
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay.
- Área I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay.
| |
Collapse
|
31
|
Wang J, Lu Y, Carr C, Dhandapani KM, Brann DW. Senolytic therapy is neuroprotective and improves functional outcome long-term after traumatic brain injury in mice. Front Neurosci 2023; 17:1227705. [PMID: 37575310 PMCID: PMC10416099 DOI: 10.3389/fnins.2023.1227705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Chronic neuroinflammation can exist for months to years following traumatic brain injury (TBI), although the underlying mechanisms remain poorly understood. Methods In the current study, we used a controlled cortical impact mouse model of TBI to examine whether proinflammatory senescent cells are present in the brain long-term (months) after TBI and whether ablation of these cells via administration of senolytic drugs can improve long-term functional outcome after TBI. The results revealed that astrocytes and microglia in the cerebral cortex, hippocampus, corpus callosum and lateral posterior thalamus colocalized the senescent cell markers, p16Ink4a or p21Cip1/Waf1 at 5 weeks post injury (5wpi) and 4 months post injury (4mpi) in a controlled cortical impact (CCI) model. Intermittent administration of the senolytic drugs, dasatinib and quercetin (D + Q) beginning 1-month after TBI for 13 weeks significantly ablated p16Ink4a-positive- and p21Cip1/Waf1-positive-cells in the brain of TBI animals, and significantly reduced expression of the major senescence-associated secretory phenotype (SASP) pro-inflammatory factors, interleukin-1β and interleukin-6. Senolytic treatment also significantly attenuated neurodegeneration and enhanced neuron number at 18 weeks after TBI in the ipsilateral cortex, hippocampus, and lateral posterior thalamus. Behavioral testing at 18 weeks after TBI further revealed that senolytic therapy significantly rescued defects in spatial reference memory and recognition memory, as well as depression-like behavior in TBI mice. Discussion Taken as a whole, these findings indicate there is robust and widespread induction of senescent cells in the brain long-term after TBI, and that senolytic drug treatment begun 1-month after TBI can efficiently ablate the senescent cells, reduce expression of proinflammatory SASP factors, reduce neurodegeneration, and rescue defects in reference memory, recognition memory, and depressive behavior.
Collapse
Affiliation(s)
| | | | | | | | - Darrell W. Brann
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
32
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
33
|
Huang Y, Sun W, Gao F, Ma H, Yuan T, Liu Z, Liu H, Hu J, Bai J, Zhang X, Wang R. Brain-Derived Estrogen Regulates Neurogenesis, Learning and Memory with Aging in Female Rats. BIOLOGY 2023; 12:760. [PMID: 37372046 DOI: 10.3390/biology12060760] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023]
Abstract
Although 17β-estradiol (E2) can be locally synthesized in the brain, whether and how brain-derived E2 (BDE2) impacts neurogenesis with aging is largely unclear. In this study, we examined the hippocampal neural stem cells, neurogenesis, and gliogenesis of 1, 3, 6, 14, and 18-month (Mon) female rats. Female forebrain neuronal aromatase knockout (FBN-ARO-KO) rats and letrozole-treated rats were also employed. We demonstraed that (1) the number of neural stem cells declined over 14-Mon age, and the differentiation of astrocytes and microglia markedly elevated and exhibited excessive activation. KO rats showed declines in astrocyte A2 subtype and elevation in A1 subtype at 18 Mon; (2) neurogenesis sharply dropped from 1-Mon age; (3) KO suppressed dentate gyrus (DG) neurogenesis at 1, 6 and 18 Mon. Additionally, KO and letrozole treatment led to declined neurogenesis at 1-Mon age, compared to age-matched WT controls; (4) FBN-ARO-KO inhibited CREB-BDNF activation, and decreased protein levels of neurofilament, spinophilin and PSD95. Notably, hippocampal-dependent spatial learning and memory was impaired in juvenile (1 Mon) and adulthood (6 Mon) KO rats. Taken together, we demonstrated that BDE2 plays a pivotal role for hippocampal neurogenesis, as well as learning and memory during female aging, especially in juvenile and middle age.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Wuxiang Sun
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Haoran Ma
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Tao Yuan
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Zixuan Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Huiyu Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Jiewei Hu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Xin Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
34
|
Zhong X, Sun Y, Lu Y, Xu L. Immunomodulatory role of estrogen in ischemic stroke: neuroinflammation and effect of sex. Front Immunol 2023; 14:1164258. [PMID: 37180115 PMCID: PMC10167039 DOI: 10.3389/fimmu.2023.1164258] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Although estrogen is predominantly related to the maintenance of reproductive functioning in females, it mediates various physiological effects in nearly all tissues, especially the central nervous system. Clinical trials have revealed that estrogen, especially 17β-estradiol, can attenuate cerebral damage caused by an ischemic stroke. One mechanism underlying this effect of 17β-estradiol is by modulating the responses of immune cells, indicating its utility as a novel therapeutic strategy for ischemic stroke. The present review summarizes the effect of sex on ischemic stroke progression, the role of estrogen as an immunomodulator in immune reactions, and the potential clinical value of estrogen replacement therapy. The data presented here will help better understand the immunomodulatory function of estrogen and may provide a basis for its novel therapeutic use in ischemic stroke.
Collapse
Affiliation(s)
- Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yajun Lu
- Department of Internal Medicine, Sunto Women & Children’s Hospital, Jiaxing, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
35
|
Kadlecova M, Freude K, Haukedal H. Complexity of Sex Differences and Their Impact on Alzheimer's Disease. Biomedicines 2023; 11:biomedicines11051261. [PMID: 37238932 DOI: 10.3390/biomedicines11051261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Sex differences are present in brain morphology, sex hormones, aging processes and immune responses. These differences need to be considered for proper modelling of neurological diseases with clear sex differences. This is the case for Alzheimer's disease (AD), a fatal neurodegenerative disorder with two-thirds of cases diagnosed in women. It is becoming clear that there is a complex interplay between the immune system, sex hormones and AD. Microglia are major players in the neuroinflammatory process occurring in AD and have been shown to be directly affected by sex hormones. However, many unanswered questions remain as the importance of including both sexes in research studies has only recently started receiving attention. In this review, we provide a summary of sex differences and their implications in AD, with a focus on microglia action. Furthermore, we discuss current available study models, including emerging complex microfluidic and 3D cellular models and their usefulness for studying hormonal effects in this disease.
Collapse
Affiliation(s)
- Marion Kadlecova
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 C Frederiksberg, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 C Frederiksberg, Denmark
| | - Henriette Haukedal
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 C Frederiksberg, Denmark
| |
Collapse
|
36
|
Wang J, Pratap UP, Lu Y, Sareddy GR, Tekmal RR, Vadlamudi RK, Brann DW. Development and Characterization of Inducible Astrocyte-Specific Aromatase Knockout Mice. BIOLOGY 2023; 12:621. [PMID: 37106821 PMCID: PMC10135694 DOI: 10.3390/biology12040621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
17β-estradiol (E2) is produced in the brain as a neurosteroid, in addition to being an endocrine signal in the periphery. The current animal models for studying brain-derived E2 include global and conditional non-inducible knockout mouse models. The aim of this study was to develop a tamoxifen (TMX)-inducible astrocyte-specific aromatase knockout mouse line (GFAP-ARO-iKO mice) to specifically deplete the E2 synthesis enzymes and aromatase in astrocytes after their development in adult mice. The characterization of the GFAP-ARO-iKO mice revealed a specific and robust depletion in the aromatase expressions of their astrocytes and a significant decrease in their hippocampal E2 levels after a GCI. The GFAP-ARO-iKO animals were alive and fertile and had a normal general brain anatomy, with a normal astrocyte shape, intensity, and distribution. In the hippocampus, after a GCI, the GFAP-ARO-iKO animals showed a major deficiency in their reactive astrogliosis, a dramatically increased neuronal loss, and increased microglial activation. These findings indicate that astrocyte-derived E2 (ADE2) regulates the ischemic induction of reactive astrogliosis and microglial activation and is neuroprotective in the ischemic brain. The GFAP-ARO-iKO mouse models thus provide an important new model to help elucidate the roles and functions of ADE2 in the brain.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Rajeshwar R. Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Darrell W. Brann
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
37
|
Shen X, Li M, Shao K, Li Y, Ge Z. Post-ischemic inflammatory response in the brain: Targeting immune cell in ischemic stroke therapy. Front Mol Neurosci 2023; 16:1076016. [PMID: 37078089 PMCID: PMC10106693 DOI: 10.3389/fnmol.2023.1076016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
An ischemic stroke occurs when the blood supply is obstructed to the vascular basin, causing the death of nerve cells and forming the ischemic core. Subsequently, the brain enters the stage of reconstruction and repair. The whole process includes cellular brain damage, inflammatory reaction, blood–brain barrier destruction, and nerve repair. During this process, the proportion and function of neurons, immune cells, glial cells, endothelial cells, and other cells change. Identifying potential differences in gene expression between cell types or heterogeneity between cells of the same type helps to understand the cellular changes that occur in the brain and the context of disease. The recent emergence of single-cell sequencing technology has promoted the exploration of single-cell diversity and the elucidation of the molecular mechanism of ischemic stroke, thus providing new ideas and directions for the diagnosis and clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xueyang Shen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Mingming Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Kangmei Shao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Yongnan Li,
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Zhaoming Ge,
| |
Collapse
|
38
|
Huang Y, Wang J, Yue C, Wang R, Guo Q, Wang T, Wang D, Dong H, Hu Y, Tao G, Li X. An In Situ Assembled Trapping Gel Repairs Spinal Cord Injury by Capturing Glutamate and Free Calcium Ions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206229. [PMID: 36683214 DOI: 10.1002/smll.202206229] [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: 10/10/2022] [Revised: 01/01/2023] [Indexed: 06/17/2023]
Abstract
Spinal cord injury (SCI) can lead to devastating autonomic dysfunction. One of the most challenging issues for functional repair in SCI is the secondary damage caused by the increased release of glutamate and free Ca2+ from injured cells. Here, an in situ assembled trapping gel (PF-SA-GAD) is developed to sweep glutamate and Ca2+ , promoting SCI repair. The hydrogel solution is a mixture of recombinant glutamate decarboxylase 67 (rGAD67) protein, sodium alginate (SA), and pluronic F-127 (PF-127). After intrathecal administration, temperature-sensitive PF-127 promoted in situ gelation. Glutamate (Glu) is captured and decarboxylated by rGAD67 into γ-aminobutyric acid (GABA). SA reacted with the free Ca2+ to generate gellable calcium alginate. Thereby, this in situ trapping gel retarded secondary neuron injury caused by Glu and free Ca2+ during SCI. In rat models of SCI, PF-SA-GAD reduces the lesion volume and inflammatory response after SCI, restores the motor function of rats with SCI. Together, the in situ assembled trapping gel is a long-term effective and minimally invasive sweeper for the direct elimination of glutamate and Ca2+ from injury lesions and can be a novel strategy for SCI repair by preventing secondary injury.
Collapse
Affiliation(s)
- Ying Huang
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jialun Wang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, 210008, China
- Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210008, China
| | - Ran Wang
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qiyuan Guo
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Tingyu Wang
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Daojuan Wang
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, 210008, China
- Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing, 210008, China
- Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210008, China
| | - Gaojian Tao
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xihan Li
- Department of Pain, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| |
Collapse
|
39
|
Wang C, Li L. The critical role of KLF4 in regulating the activation of A1/A2 reactive astrocytes following ischemic stroke. J Neuroinflammation 2023; 20:44. [PMID: 36823628 PMCID: PMC9948409 DOI: 10.1186/s12974-023-02742-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND We have previously demonstrated that the expression of kruppel-like transcription factor-4 (KLF-4) is upregulated in astrocytes following acute ischemic stroke (AIS) and found that KLF4 confers vascular protection against cerebral ischemic injury. However, the functional role of KLF4 in astrocyte after AIS is far from clear. METHODS The intrinsic relationship between KLF4 and A1/A2 reactive astrocytes and the impact of astrocytic KLF4 on the activation of A1/A2 subtype astrocytes were evaluated in middle cerebral artery occlusion (MCAO) mice and oxygen-glucose deprivation and restoration (OGD/R) astrocytes. RESULTS Our results demonstrated that astrocytic KLF4 expression and complement C3-positive A1 and S100 calcium binding protein A10 (S100A10)-positive A2 astrocytes were induced in the ischemic penumbra following focal cerebral ischemia, and the time course of upregulation of astrocytic KLF4 correlated closely with the activation of A2 astrocytes. The dual immunofluorescent studies displayed that in the ischemic hemisphere, where the high levels of KLF4 were expressed, there were relatively low levels of C3 expressed in the reactive astrocytes and vice versa, but KLF4 was always co-stained well with S100A10. Mechanistic analyses revealed that astrocytic KLF4 inhibited the activation of A1 astrocyte but promoted A2 astrocyte polarization after OGD/R by modulating expressions of nuclear factor-kB. CONCLUSIONS Astrocyte-derived KLF4 has a critical role in regulating the activation of A1/A2 reactive astrocytes following AIS.
Collapse
Affiliation(s)
- Cong Wang
- grid.412277.50000 0004 1760 6738Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 People’s Republic of China ,grid.412194.b0000 0004 1761 9803The Graduate School, Ningxia Medical University, Yinchuan, Ningxia 750004 People’s Republic of China
| | - Longxuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
40
|
Sabetta Z, Krishna G, Curry T, Adelson PD, Thomas TC. Aging with TBI vs. Aging: 6-month temporal profiles for neuropathology and astrocyte activation converge in behaviorally relevant thalamocortical circuitry of male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527058. [PMID: 36798182 PMCID: PMC9934568 DOI: 10.1101/2023.02.06.527058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Traumatic brain injury (TBI) manifests late-onset and persisting clinical symptoms with implications for sex differences and increased risk for the development of age-related neurodegenerative diseases. Few studies have evaluated chronic temporal profiles of neuronal and glial pathology that include sex as a biological variable. After experimental diffuse TBI, late-onset and persisting somatosensory hypersensitivity to whisker stimulation develops at one-month post-injury and persists to at least two months post-injury in male rats, providing an in vivo model to evaluate the temporal profile of pathology responsible for morbidity. Whisker somatosensation is dependent on signaling through the thalamocortical relays of the whisker barrel circuit made up of glutamatergic projections between the ventral posteromedial nucleus of the thalamus (VPM) and primary somatosensory barrel cortex (S1BF) with inhibitory (GABA) innervation from the thalamic reticular nucleus (TRN) to the VPM. To evaluate the temporal profiles of pathology, male and female Sprague Dawley rats ( n = 5-6/group) were subjected to sham surgery or midline fluid percussion injury (FPI). At 7-, 56-, and 168-days post-injury (DPI), brains were processed for amino-cupric silver stain and glial fibrillary acidic protein (GFAP) immunoreactivity, where pixel density of staining was quantified to determine the temporal profile of neuropathology and astrocyte activation in the VPM, S1BF, and TRN. FPI induced significant neuropathology in all brain regions at 7 DPI. At 168 DPI, neuropathology remained significantly elevated in the VPM and TRN, but returned to sham levels in the S1BF. GFAP immunoreactivity was increased as a function of FPI and DPI, with an FPI × DPI interaction in all regions and an FPI × Sex interaction in the S1BF. The interactions were driven by increased GFAP immunoreactivity in shams over time in the VPM and TRN. In the S1BF, GFAP immunoreactivity increased at 7 DPI and declined to age-matched sham levels by 168 DPI, while GFAP immunoreactivity in shams significantly increased between 7 and 168 days. The FPI × Sex interaction was driven by an overall greater level of GFAP immunoreactivity in FPI males compared to FPI females. Increased GFAP immunoreactivity was associated with an increased number of GFAP-positive soma, predominantly at 7 DPI. Overall, these findings indicate that FPI, time post-injury, sex, region, and aging with injury differentially contribute to chronic changes in neuronal pathology and astrocyte activation after diffuse brain injury. Thus, our results highlight distinct patterns of pathological alterations associated with the development and persistence of morbidity that supports chronic neuropathology, especially within the thalamus. Further, data indicate a convergence between TBI-induced and age-related pathology where further investigation may reveal a role for divergent astrocytic phenotypes associated with increased risk for neurodegenerative diseases.
Collapse
|
41
|
Saldanha CJ. Spatial and temporal specificity of neuroestradiol provision in the songbird. J Neuroendocrinol 2023; 35:e13192. [PMID: 35983989 PMCID: PMC9889572 DOI: 10.1111/jne.13192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 02/03/2023]
Abstract
Steroid hormones are often synthesized in multiple tissues, affect several different targets, and modulate numerous physiological endpoints. The mechanisms by which this modulation is achieved with temporal and spatial specificity remain unclear. 17β-estradiol for example, is made in several peripheral tissues and in the brain, where it affects a diverse set of behaviors. How is estradiol delivered to the right target, at the right time, and at the right concentration? In the last two decades, we have learned that aromatase (estrogen-synthase) can be induced in astrocytes following damage to the brain and is expressed at central synapses. Both mechanisms of estrogen provision confer spatial and temporal specificity on a lipophilic neurohormone with potential access to all cells and tissues. In this review, I trace the progress in our understanding of astrocytic and synaptic aromatization. I discuss the incidence, regulation, and functions of neuroestradiol provision by aromatization, first in astrocytes and then at synapses. Finally, I focus on a relatively novel hypothesis about the role of neuroestradiol in the orchestration of species-specific behaviors.
Collapse
Affiliation(s)
- Colin J Saldanha
- Departments of Neuroscience and Psychology, and Center for Behavioral Neuroscience, American University, Washington, DC, USA
| |
Collapse
|
42
|
Abstract
Stroke is a sudden and rapidly progressing ischemic or hemorrhagic cerebrovascular disease. When stroke damages the brain, the immune system becomes hyperactive, leading to systemic inflammatory response and immunomodulatory disorders, which could significantly impact brain damage, recovery, and prognosis of stroke. Emerging researches suggest that ischemic stroke-induced spleen contraction could activate a peripheral immune response, which may further aggravate brain injury. This review focuses on hemorrhagic strokes including intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH) and discusses the central nervous system-peripheral immune interactions after hemorrhagic stroke induction. First, inflammatory progression after ICH and SAH is investigated. As a part of this review, we summarize the various kinds of inflammatory cell infiltration to aggravate brain injury after blood-brain barrier interruption induced by hemorrhagic stroke. Then, we explore hemorrhagic stroke-induced systemic inflammatory response syndrome (SIRS) and discuss the interactions of CNS and peripheral inflammatory response. In addition, potential targets related to inflammatory response for ICH and SAH are discussed in this review, which may lead to novel therapeutic strategies for hemorrhagic stroke.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Stroke Research, Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Stroke Research, Soochow University, Suzhou, China
| |
Collapse
|
43
|
Anti-Inflammatory Actions of G-Protein-Coupled Estrogen Receptor 1 (GPER) and Brain-Derived Estrogen Following Cerebral Ischemia in Ovariectomized Rats. BIOLOGY 2023; 12:biology12010099. [PMID: 36671793 PMCID: PMC9855882 DOI: 10.3390/biology12010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Global cerebral ischemia can elicit rapid innate neuroprotective mechanisms that protect against delayed neuronal death. Brain-derived 17β-estradiol (BDE2), an endogenous neuroprotectant, is synthesized from testosterone by the enzyme aromatase (Aro) and is upregulated by brain ischemia and inflammation. Our recent study revealed that G1, a specific G-protein-coupled estrogen receptor 1 (GPER) agonist, exerts anti-inflammatory and anti-apoptotic roles after global cerebral ischemia (GCI). Herein, we aimed to elucidate whether G1 modulates the early inflammatory process and the potential underlying mechanisms in the ovariectomized rat hippocampal CA1 region. G1 was found to markedly reduce pro-inflammatory (iNOS, MHCII, and CD68) and to enhance anti-inflammatory (CD206, Arginase 1, IL1RA, PPARγ, and BDNF) markers after 1 and 3 days of reperfusion after GCI. Intriguingly, the neuroprotection of G1 was blocked by the Aro inhibitor, letrozole. Conversely, the GPER antagonist, G36, inhibited Aro-BDE2 signaling and exacerbated neuronal damage. As a whole, this work demonstrates a novel anti-inflammatory role of GPER, involving a synergistic mediation with BDE2 during the early stage of GCI.
Collapse
|
44
|
Hasegawa H, Kondo M. Astrocytic Responses to Binge Alcohol Intake in the Mouse Hindbrain. Biol Pharm Bull 2023; 46:1194-1202. [PMID: 37661398 DOI: 10.1248/bpb.b23-00140] [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] [Indexed: 09/05/2023]
Abstract
Ethanol is the most commonly used toxic chemical in human cultures. Ethanol predominantly damages the brain causing various neurological disorders. Astrocytes are important cellular targets of ethanol in the brain and are involved in alcoholic symptoms. Recent studies have revealed the diversity of astrocyte populations in the brain. However, it is unclear how the different astrocyte populations respond to an excess of ethanol. Here we examined the effect of binge ethanol levels on astrocytes in the mouse brainstem and cerebellum. Ethanol administration for four consecutive days increased the glial fibrillary acidic protein (GFAP)-immunoreactive signals in the spinal tract of the trigeminal nerve (stTN) and reticular nucleus (RN). Another astrocyte marker, aquaporin 4 (AQP4), was also increased in the stTN with a pattern similar to that of GFAP. However, in the RN, the immunoreactive signals of AQP4 were different from that of GFAP and were not changed by ethanol administration. In the cerebellum, GFAP-positive signals were found in all four astrocytic populations, and those in the Bergmann glia were selectively eliminated by ethanol administration. We next examined the effect of estradiol on the ethanol-induced changes in astrocytic immunoreactive signals. The administration of estradiol alone increased the AQP4-immunoreactivity in the stTN with a pattern similar to that of ethanol, whereas the co-administration of estradiol and ethanol suppressed the intensity of the AQP4-positive signals. Thus, binge levels of ethanol intake selectively affect astrocyte populations in the brainstem and cerebellum. Sex hormones can affect the ethanol-induced neurotoxicity via modulation of astrocyte reactivity.
Collapse
Affiliation(s)
| | - Mari Kondo
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University
| |
Collapse
|
45
|
Zhang R, Chen M, Deng Z, Kong L, Shen B, Zhang L. Delta Opioid Peptide Targets Brain Microvascular Endothelial Cells Reducing Apoptosis to Relieve Hypoxia-Ischemic/Reperfusion Injury. Pharmaceutics 2022; 15:pharmaceutics15010046. [PMID: 36678674 PMCID: PMC9861451 DOI: 10.3390/pharmaceutics15010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is one of the leading causes of death. (D-ala2, D-leu5) enkephalin (DADLE) is a synthetic peptide and highly selective delta opioid receptor (δOR) agonist that has exhibited protective properties in ischemia. However, the specific target and mechanism are still unclear. The present study explores the expression of δOR on brain microvascular endothelial cells (BMECs) and whether DADLE could relieve I/R-induced injury by reducing apoptosis. A lateral ventricular injection of DADLE for pretreatment, the neurofunctional behavior score, and TTC staining, were used to evaluate the protective effect of DADLE. Immunofluorescence technology was used to label different types of cells with apoptosis-positive signals to test co-localization status. Primary cultured BMECs were separated and treated with DADLE, accompanied by OGD/R. The CCK-8 test was conducted to evaluate cell viability and TdT-mediated dUTP Nick-end Labelling (TUNEL) staining to test apoptosis levels. The levels of apoptosis-related proteins were analyzed by Western blotting. The co-localization results showed that BMECs, but not astrocytes, microglia, or neurons, presented mostly TUNEL-positive signals, especially in the Dentate gyrus (DG) area of the hippocampus. Either activation of δORs on rats' brains or primary BMECs mainly reduce cellular apoptosis and relieve the injury. Interference with the expression δOR could block this effect. DADLE also significantly increased levels of Bcl-2 and reduced levels of Bax. δOR's expressions can be detected on the BMECs, but not on the HEK293 cells, by Western blotting and IFC. Therefore, DADLE exerts a cytoprotective effect, primarily under hypoxia-ischemic injury/reperfusion conditions, by targeting BMECs to inhibit apoptosis.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Meixuan Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongfang Deng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Bing Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Correspondence:
| |
Collapse
|
46
|
Zhang Y, Zhao X, Zhang Y, Zeng F, Yan S, Chen Y, Li Z, Zhou D, Liu L. The role of circadian clock in astrocytes: From cellular functions to ischemic stroke therapeutic targets. Front Neurosci 2022; 16:1013027. [PMID: 36570843 PMCID: PMC9772621 DOI: 10.3389/fnins.2022.1013027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests that astrocytes, the abundant cell type in the central nervous system (CNS), play a critical role in maintaining the immune response after cerebral infarction, regulating the blood-brain barrier (BBB), providing nutrients to the neurons, and reuptake of glutamate. The circadian clock is an endogenous timing system that controls and optimizes biological processes. The central circadian clock and the peripheral clock are consistent, controlled by various circadian components, and participate in the pathophysiological process of astrocytes. Existing evidence shows that circadian rhythm controls the regulation of inflammatory responses by astrocytes in ischemic stroke (IS), regulates the repair of the BBB, and plays an essential role in a series of pathological processes such as neurotoxicity and neuroprotection. In this review, we highlight the importance of astrocytes in IS and discuss the potential role of the circadian clock in influencing astrocyte pathophysiology. A comprehensive understanding of the ability of the circadian clock to regulate astrocytes after stroke will improve our ability to predict the targets and biological functions of the circadian clock and gain insight into the basis of its intervention mechanism.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yao Chen
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhong Li
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,Desheng Zhou,
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,*Correspondence: Lijuan Liu,
| |
Collapse
|
47
|
Kim SC, Park JY, Hwang EM. Caspase-dependent apoptosis induces reactivation and gliogenesis of astrocytes in adult mice. Front Cell Neurosci 2022; 16:1054956. [DOI: 10.3389/fncel.2022.1054956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Astrocytes play an important role in increasing synaptic plasticity, regulating endogenous homeostasis, and contributing to neuroprotection but become overactivated or apoptotic in persistent neuroinflammatory responses or pathological conditions. Although gliogenesis under these conditions may be essential for neuronal protection, much remains unknown. Here, we generated new conditional transgenic mice (cTg) that can induce apoptosis via Cre-dependent active caspase-3 (taCasp3-2A-TEVp) without pathological conditions. We induced apoptosis of hippocampal CA1 astrocytes in cTg mice using GFAP promoter-driven adeno-associated virus (AAV) containing Cre recombinase. Activated caspase-3 was detected in astrocytes of the hippocampal CA1, and the number of astrocytes decreased sharply at 1 week but recovered at 2 weeks and was maintained until 4 weeks. Nuclear factor 1A (NF1A) mRNA, an important transcription factor for hippocampal reactive astrocytes, was significantly increased only at week 1. Interestingly, all reactive markers (pan, A1, A2) increased despite the decreased number of astrocytes at week 1, and there was no change in monoamine oxidase B (MAOB) observed in astrocytes of animal models of degenerative brain disease. Extensive CA1 astrocyte depletion at week 1 induced cognitive deficits; however, both recovered at weeks 2 and 4. Overall, transient hippocampal astrocyte depletion caused by apoptosis restored cell number and function within 2 weeks and did not induce significant neurotoxicity. Therefore, cTg mice are valuable as an in vivo animal model for studying gliogenesis in multiple regions of the adult brain.
Collapse
|
48
|
Ding Z, Li F, Xie L, Gu M, Li C, Liu C, Peng C, Li W. Design and Synthesis of Novel Phenylahistin Derivatives Based on Co-Crystal Structures as Potent Microtubule Inhibitors for Anti-Cancer Therapy. Mar Drugs 2022; 20:752. [PMID: 36547899 PMCID: PMC9785606 DOI: 10.3390/md20120752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022] Open
Abstract
Phenylahistin is a naturally occurring marine product with a diketopiperazine structure that can bind to the colchicine site of microtubulin as a possible anticancer agent. To develop more potent microtubule inhibitors, novel phenylahistin derivatives were designed and synthesized based on the co-crystal complexes of phenylahistin derivatives and microtubulin. We established a focused library of imidazole-type molecules for the introduction of different groups to the C-ring and A-ring of phenylahistin. Structure-activity relationship studies indicated that appropriate hydrocarbon substituents and unsaturated alkenyl substituents at the 1-position of the imidazole group are important for improving the activity of such compounds. In addition, this study found that propylamine groups could maintain the activity of these compounds, as exemplified by compound 16d (IC50 = 5.38 nM, NCI-H460). Compound 15p (IC50 = 1.03 nM, NCI-H460) with an allyl group exhibited potent cytotoxic activity at the nanomolar level against human lung cancer cell lines. Immunofluorescence assay indicated that compound 15p could efficiently inhibited microtubule polymerization and induced a high expression of caspase-3. 15p also displayed good pharmacokinetic characteristics in vitro. Additionally, the growth of H22 transplanted tumors was significantly inhibited in BALB/c mice when 15p alone was administered at 4 mg/kg, and the tumor inhibition rate was as much as 65%. Importantly, the continuous administration of 15p resulted in a lower toxicity than that of docetaxel (10 mg/kg) and cyclophosphamide (20 mg/kg). Overall, the novel allyl-imidazole-diketopiperazine-type derivatives could be considered safe and effective potential agents for cancer treatment.
Collapse
Affiliation(s)
- Zhongpeng Ding
- Medical College, Linyi University, Shuangling Road, Linyi 276000, China
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
| | - Feifei Li
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
| | - Lianghui Xie
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
| | - Minqing Gu
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
| | - Chunlei Li
- Medical College, Linyi University, Shuangling Road, Linyi 276000, China
| | - Chang Liu
- Medical College, Linyi University, Shuangling Road, Linyi 276000, China
| | - Chao Peng
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
| | - Wenbao Li
- Shenzhen BGI Marine Science & Technology Co., Ltd., Shenzhen 518002, China
- Shenzhen Huahong Marine Biomedical Co., Ltd., Shenzhen 518002, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Innovation Center for Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| |
Collapse
|
49
|
Brain-Derived Estrogen and Neurological Disorders. BIOLOGY 2022; 11:biology11121698. [PMID: 36552208 PMCID: PMC9774965 DOI: 10.3390/biology11121698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Astrocytes and neurons in the male and female brains produce the neurosteroid brain-derived 17β-estradiol (BDE2) from androgen precursors. In this review, we discuss evidence that suggest BDE2 has a role in a number of neurological conditions, such as focal and global cerebral ischemia, traumatic brain injury, excitotoxicity, epilepsy, Alzheimer's disease, and Parkinson's disease. Much of what we have learned about BDE2 in neurological disorders has come from use of aromatase inhibitors and global aromatase knockout mice. Recently, our group developed astrocyte- and neuron-specific aromatase knockout mice, which have helped to clarify the precise functions of astrocyte-derived 17β-estradiol (ADE2) and neuron-derived 17β-estradiol (NDE2) in the brain. The available evidence to date suggests a primarily beneficial role of BDE2 in facilitating neuroprotection, synaptic and cognitive preservation, regulation of reactive astrocyte and microglia activation, and anti-inflammatory effects. Most of these beneficial effects appear to be due to ADE2, which is induced in most neurological disorders, but there is also recent evidence that NDE2 exerts similar beneficial effects. Furthermore, in certain situations, BDE2 may also have deleterious effects, as recent evidence suggests its overproduction in epilepsy contributes to seizure induction. In this review, we examine the current state of this quickly developing topic, as well as possible future studies that may be required to provide continuing growth in the field.
Collapse
|
50
|
Yang R, Pan J, Wang Y, Xia P, Tai M, Jiang Z, Chen G. Application and prospects of somatic cell reprogramming technology for spinal cord injury treatment. Front Cell Neurosci 2022; 16:1005399. [PMID: 36467604 PMCID: PMC9712200 DOI: 10.3389/fncel.2022.1005399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a serious neurological trauma that is challenging to treat. After SCI, many neurons in the injured area die due to necrosis or apoptosis, and astrocytes, oligodendrocytes, microglia and other non-neuronal cells become dysfunctional, hindering the repair of the injured spinal cord. Corrective surgery and biological, physical and pharmacological therapies are commonly used treatment modalities for SCI; however, no current therapeutic strategies can achieve complete recovery. Somatic cell reprogramming is a promising technology that has gradually become a feasible therapeutic approach for repairing the injured spinal cord. This revolutionary technology can reprogram fibroblasts, astrocytes, NG2 cells and neural progenitor cells into neurons or oligodendrocytes for spinal cord repair. In this review, we provide an overview of the transcription factors, genes, microRNAs (miRNAs), small molecules and combinations of these factors that can mediate somatic cell reprogramming to repair the injured spinal cord. Although many challenges and questions related to this technique remain, we believe that the beneficial effect of somatic cell reprogramming provides new ideas for achieving functional recovery after SCI and a direction for the development of treatments for SCI.
Collapse
Affiliation(s)
- Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Yankai Wang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Panhui Xia
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Mingliang Tai
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Zhihao Jiang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|