1
|
Sharma V, Singh TG. Hypoxia-inducible Factor-1α Pathway in Cerebral Ischemia: From Molecular Mechanisms to Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:208-218. [PMID: 39428931 DOI: 10.2174/0118715273324551241008111827] [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: 05/31/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Ischemic injury to the brain can result in a variety of life-threatening conditions, mortality, or varying degrees of disability. Hypoxia-inducible factor 1α (HIF 1α), an oxygen- sensitive transcription factor that controls the adaptive metabolic response to hypoxia, is a critical constituent of cerebral ischemia. It participates in numerous processes, such as metabolism, proliferation, and angiogenesis, and plays a major role in cerebral ischemia. METHODS Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the pharmacological modulation of HIF-1α pathways for the treatment of cerebral ischemia. RESULTS Various signalling pathways, such as Mitogen-activated protein kinase (MAPK), Janus kinase/ signal transducers and activators (JAK/STAT), Phosphoinositide-3-kinase (PI3-K), and cAMPresponse element binding protein (CREB) play a vital role in modulation of HIF-1α pathway, which helps in preventing the pathogenesis of cerebral ischemia. CONCLUSION The pharmacological modulation of the HIF-1α pathway via various molecular signalling pathways, such as PI3-K, MAPK, CREB, and JAK/STAT activators, offer a promising prospect for future interventions and treatment for cerebral ischemia.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
2
|
Exploring Active Compounds and Mechanisms of Angong Niuhuang Wan on Ischemic Stroke Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2443615. [PMID: 35388303 PMCID: PMC8977296 DOI: 10.1155/2022/2443615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/23/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Methods The chemical ingredients of ANW were retrieved from TCMSP, TCMID, and literature. We predicted the potential targets of active ingredients by PubChem, Swiss Target Prediction, and STITCH databases. The targets related to ischemic stroke were retrieved using GeneCards, DisGeNET, DrugBank, TTD, and GEO databases. Subsequently, Venn diagrams were used to identify common targets of active ingredients and ischemic stroke. Protein-protein interaction (PPI) network was structured with STRING platform and Cytoscape 3.8.2. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of key targets were performed in the Metascape database. Finally, molecular docking was conducted by AutoDock Tools and PyMOL software. Results A total of 2391 targets were identified for 230 active ingredients of ANW, and 1386 of them overlapped with ischemic stroke targets. The key active ingredients were mainly quercetin, β-estradiol, berberine, wogonin, and β-sitosterol, and the key targets were also identified, including IL-6, AKT1, MAPK3, PIK3CA, and TNF. The biological process (BP) results indicated that ANW may have therapeutic effects through response oxidative stress, inflammatory response, cellular response to lipid, and response to nutrient levels. Furthermore, the ingredients of ANW were predicted to have therapeutic effects on ischemic stroke via the HIF-1 signaling pathway, FoxO signaling pathway, chemokine signaling pathway, fluid shear stress and atherosclerosis, and neurotrophin signaling pathway. The molecular docking results all showed that the core ingredients were strong binding activity with the core targets. Conclusion In conclusion, the bioinformatics and pharmacological results reveal that counteracting oxidative stress, suppressing inflammation, inhibiting the development of AS, and even protecting neurological function are critical pathways for ANW in the treatment of ischemic stroke. These results may help to elucidate the mechanism of ANW on ischemic stroke for experimental studies and clinical applications.
Collapse
|
3
|
Premarin Reduces Neurodegeneration and Promotes Improvement of Function in an Animal Model of Spinal Cord Injury. Int J Mol Sci 2022; 23:ijms23042384. [PMID: 35216504 PMCID: PMC8875481 DOI: 10.3390/ijms23042384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023] Open
Abstract
Spinal cord injury (SCI) causes significant mortality and morbidity. Currently, no FDA-approved pharmacotherapy is available for treating SCI. Previously, low doses of estrogen (17β-estradiol, E2) were shown to improve the post-injury outcome in a rat SCI model. However, the range of associated side effects makes advocating its therapeutic use difficult. Therefore, this study aimed at investigating the therapeutic efficacy of Premarin (PRM) in SCI. PRM is an FDA-approved E2 (10%) formulation, which is used for hormone replacement therapy with minimal risk of serious side effects. The effects of PRM on SCI were examined by magnetic resonance imaging, immunofluorescent staining, and western blot analysis in a rat model. SCI animals treated with vehicle alone, PRM, E2 receptor antagonist (ICI), or PRM + ICI were graded in a blinded way for locomotor function by using the Basso–Beattie–Bresnahan (BBB) locomotor scale. PRM treatment for 7 days decreased post-SCI lesion volume and attenuated neuronal cell death, inflammation, and axonal damage. PRM also altered the balance of pro- and anti-apoptotic proteins in favor of cell survival and improved angiogenesis and microvascular growth. Increased expression of estrogen receptors (ERs) ERα and ERβ following PRM treatment and their inhibition by ER inhibitor indicated that the neuroprotection associated with PRM treatment might be E2-receptor mediated. The attenuation of glial activation with decreased inflammation and cell death, and increased angiogenesis by PRM led to improved functional outcome as determined by the BBB locomotor scale. These results suggest that PRM treatment has significant therapeutic implications for the improvement of post-SCI outcome.
Collapse
|
4
|
Revealing the Influences of Sex Hormones and Sex Differences in Atrial Fibrillation and Vascular Cognitive Impairment. Int J Mol Sci 2021; 22:ijms22168776. [PMID: 34445515 PMCID: PMC8396287 DOI: 10.3390/ijms22168776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
The impacts of sex differences on the biology of various organ systems and the influences of sex hormones on modulating health and disease have become increasingly relevant in clinical and biomedical research. A growing body of evidence has recently suggested fundamental sex differences in cardiovascular and cognitive function, including anatomy, pathophysiology, incidence and age of disease onset, symptoms affecting disease diagnosis, disease severity, progression, and treatment responses and outcomes. Atrial fibrillation (AF) is currently recognized as the most prevalent sustained arrhythmia and might contribute to the pathogenesis and progression of vascular cognitive impairment (VCI), including a range of cognitive deficits, from mild cognitive impairment to dementia. In this review, we describe sex-based differences and sex hormone functions in the physiology of the brain and vasculature and the pathophysiology of disorders therein, with special emphasis on AF and VCI. Deciphering how sex hormones and their receptor signaling (estrogen and androgen receptors) potentially impact on sex differences could help to reveal disease links between AF and VCI and identify therapeutic targets that may lead to potentially novel therapeutic interventions early in the disease course of AF and VCI.
Collapse
|
5
|
Zheng J, Wei Z, Yang K, Lu Y, Lu P, Zhao J, Du Y, Zhang H, Li R, Lei S, Lv H, Chen X, Liu Y, Chen YM, Zhang Q, Zhang P. Neural Stem Cell-Laden Self-Healing Polysaccharide Hydrogel Transplantation Promotes Neurogenesis and Functional Recovery after Cerebral Ischemia in Rats. ACS APPLIED BIO MATERIALS 2021; 4:3046-3054. [PMID: 35014393 DOI: 10.1021/acsabm.0c00934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Exploring a strategy to effectively repair cerebral ischemic injury is a critical requirement for neuroregeneration. Herein, we transplanted a neural stem cell (NSC)-laden self-healing and injectable hydrogel into the brains of ischemic rats and evaluated its therapeutic effects. We observed an improvement in neurological functions in rats transplanted with the NSC-laden hydrogel. This strategy is sufficiently efficient to support neuroregeneration evidenced by NSC proliferation, differentiation, and athletic movement recovery of rats. This therapeutic effect relates to the inhibition of the astrocyte reaction and the increased expression of vascular endothelial growth factor. This work provides a novel approach to repair cerebral ischemic injury.
Collapse
Affiliation(s)
- Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Zhao Wei
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Kuan Yang
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Jingyi Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Yin Du
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Yong Mei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Qiqing Zhang
- Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen 518020, P. R. China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| |
Collapse
|
6
|
Josiah SS, Meor Azlan NF, Zhang J. Targeting the WNK-SPAK/OSR1 Pathway and Cation-Chloride Cotransporters for the Therapy of Stroke. Int J Mol Sci 2021; 22:1232. [PMID: 33513812 PMCID: PMC7865768 DOI: 10.3390/ijms22031232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/05/2023] Open
Abstract
Stroke is one of the major culprits responsible for morbidity and mortality worldwide, and the currently available pharmacological strategies to combat this global disease are scanty. Cation-chloride cotransporters (CCCs) are expressed in several tissues (including neurons) and extensively contribute to the maintenance of numerous physiological functions including chloride homeostasis. Previous studies have implicated two CCCs, the Na+-K+-Cl- and K+-Cl- cotransporters (NKCCs and KCCs) in stroke episodes along with their upstream regulators, the with-no-lysine kinase (WNKs) family and STE20/SPS1-related proline/alanine rich kinase (SPAK) or oxidative stress response kinase (OSR1) via a signaling pathway. As the WNK-SPAK/OSR1 pathway reciprocally regulates NKCC and KCC, a growing body of evidence implicates over-activation and altered expression of NKCC1 in stroke pathology whilst stimulation of KCC3 during and even after a stroke event is neuroprotective. Both inhibition of NKCC1 and activation of KCC3 exert neuroprotection through reduction in intracellular chloride levels and thus could be a novel therapeutic strategy. Hence, this review summarizes the current understanding of functional regulations of the CCCs implicated in stroke with particular focus on NKCC1, KCC3, and WNK-SPAK/OSR1 signaling and discusses the current and potential pharmacological treatments for stroke.
Collapse
Affiliation(s)
| | | | - Jinwei Zhang
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter EX4 4PS, UK; (S.S.J.); (N.F.M.A.)
| |
Collapse
|
7
|
Li W, Lu P, Lu Y, Wei H, Niu X, Xu J, Wang K, Zhang H, Li R, Qiu Z, Wang N, Jia P, Zhang Y, Zhang S, Lu H, Chen X, Liu Y, Zhang P. 17β-Estradiol Protects Neural Stem/Progenitor Cells Against Ketamine-Induced Injury Through Estrogen Receptor β Pathway. Front Neurosci 2020; 14:576813. [PMID: 33100963 PMCID: PMC7556164 DOI: 10.3389/fnins.2020.576813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Ketamine inhibits neural stem/progenitor cell (NSPC) proliferation and disrupts normal neurogenesis in the developing brain. 17β-Estradiol alleviates neurogenesis damage and enhances behavioral performance after ketamine administration. However, the receptor pathway of 17β-estradiol that protects NSPCs from ketamine-induced injury remains unknown. In the present study, we investigated the role of estrogen receptor α (ER-α) and estrogen receptor β (ER-β) in 17β-estradiol’s protection against ketamine-exposed NSPCs and explored its potential mechanism. The primary cultured NSPCs were identified by immunofluorescence and then treated with ketamine and varying doses of ER-α agonist 4,4′,4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT) or ER-β agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) for 24 h. NSPC proliferation was analyzed by 5-bromo-2-deoxyuridine incorporation test. The expression of phosphorylated glycogen synthase kinase-3β (p-GSK-3β) was quantified by western blotting. It was found that treatment with different concentrations of PPT did not alter the inhibition of ketamine on NSPC proliferation. However, treatment with DPN attenuated the inhibition of ketamine on NSPC proliferation at 24 h after their exposure (P < 0.05). Furthermore, treatment with DPN increased p-GSK-3β expression in NSPCs exposed to ketamine. These findings indicated that ER-β mediates probably the protective effects of 17β-estradiol on ketamine-damaged NSPC proliferation and GSK-3β is involved in this process
Collapse
Affiliation(s)
- Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Niu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing Xu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shuyue Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Azcoitia I, Barreto GE, Garcia-Segura LM. Molecular mechanisms and cellular events involved in the neuroprotective actions of estradiol. Analysis of sex differences. Front Neuroendocrinol 2019; 55:100787. [PMID: 31513774 DOI: 10.1016/j.yfrne.2019.100787] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022]
Abstract
Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland.
| | - Luis M Garcia-Segura
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain; Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| |
Collapse
|
9
|
Li W, Li H, Wei H, Lu Y, Lei S, Zheng J, Lu H, Chen X, Liu Y, Zhang P. 17β-Estradiol Treatment Attenuates Neurogenesis Damage and Improves Behavior Performance After Ketamine Exposure in Neonatal Rats. Front Cell Neurosci 2019; 13:251. [PMID: 31263401 PMCID: PMC6585163 DOI: 10.3389/fncel.2019.00251] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 05/20/2019] [Indexed: 12/23/2022] Open
Abstract
Ketamine exposure disturbed normal neurogenesis in the developing brain and resulted in subsequent neurocognitive deficits. 17β-estradiol provides robust neuroprotection in a variety of brain injury models in animals of both sexes and attenuates neurodegeneration induced by anesthesia agents. In the present study, we aimed to investigate whether 17β-estradiol could attenuate neonatal ketamine exposure-disturbed neurogenesis and behavioral performance. We treated 7-day-old (Postnatal day 7, PND 7) Sprague-Dawley rats and neural stem cells (NSCs) with either normal saline, ketamine, or 17β-estradiol before/after ketamine exposure, respectively. At PND 14, the rats were decapitated to detect neurogenesis in the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampus by immunofluorescence staining. The proliferation, neuronal differentiation, and apoptosis of NSCs were assessed by immunohistochemistry method and TUNEL assay, respectively. The protein levels of cleaved caspase-3 in vivo in addition to GSK-3β and p-GSK-3β in vitro were examined by western blotting. Spatial learning and memory abilities were assessed by Morris water maze (MWM) test at PND 42–47. Ketamine exposure decreased cell proliferation in the SVZ and SGZ, inhibited NSC proliferation and neuronal differentiation, promoted NSC apoptosis and led to adult cognitive deficits. Furthermore, ketamine increased cleaved caspase-3 in vivo and decreased the expression of p-GSK-3β in vitro. Treatment with 17β-estradiol could attenuate ketamine-induced changes both in vivo and in vitro. For the first time we showed that 17β-estradiol alleviated ketamine-induced neurogenesis inhibition and cognitive dysfunction in the developing rat brain. Moreover, the protection of 17β-estradiol was associated with GSK-3β.
Collapse
Affiliation(s)
- Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huixian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Robison LS, Gannon OJ, Salinero AE, Zuloaga KL. Contributions of sex to cerebrovascular function and pathology. Brain Res 2018; 1710:43-60. [PMID: 30580011 DOI: 10.1016/j.brainres.2018.12.030] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
Sex differences exist in how cerebral blood vessels function under both physiological and pathological conditions, contributing to observed sex differences in risk and outcomes of cerebrovascular diseases (CBVDs), such as vascular contributions to cognitive impairment and dementia (VCID) and stroke. Throughout most of the lifespan, women are protected from CBVDs; however, risk increases following menopause, suggesting sex hormones may play a significant role in this protection. The cerebrovasculature is a target for sex hormones, including estrogens, progestins, and androgens, where they can influence numerous vascular functions and pathologies. While there is a plethora of information on estrogen, the effects of progestins and androgens on the cerebrovasculature are less well-defined. Estrogen decreases cerebral tone and increases cerebral blood flow, while androgens increase tone. Both estrogens and androgens enhance angiogenesis/cerebrovascular remodeling. While both estrogens and androgens attenuate cerebrovascular inflammation, pro-inflammatory effects of androgens under physiological conditions have also been demonstrated. Sex hormones exert additional neuroprotective effects by attenuating oxidative stress and maintaining integrity and function of the blood brain barrier. Most animal studies utilize young, healthy, gonadectomized animals, which do not mimic the clinical conditions of aging individuals likely to get CBVDs. This is also concerning, as sex hormones appear to mediate cerebrovascular function differently based on age and disease state (e.g. metabolic syndrome). Through this review, we hope to inspire others to consider sex as a key biological variable in cerebrovascular research, as greater understanding of sex differences in cerebrovascular function will assist in developing personalized approaches to prevent and treat CBVDs.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| |
Collapse
|
11
|
Gannon OJ, Robison LS, Custozzo AJ, Zuloaga KL. Sex differences in risk factors for vascular contributions to cognitive impairment & dementia. Neurochem Int 2018; 127:38-55. [PMID: 30471324 DOI: 10.1016/j.neuint.2018.11.014] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is the second most common cause of dementia. While males overall appear to be at a slightly higher risk for VCID throughout most of the lifespan (up to age 85), some risk factors for VCID more adversely affect women. These include female-specific risk factors associated with pregnancy related disorders (e.g. preeclampsia), menopause, and poorly timed hormone replacement. Further, presence of certain co-morbid risk factors, such as diabetes, obesity and hypertension, also may more adversely affect women than men. In contrast, some risk factors more greatly affect men, such as hyperlipidemia, myocardial infarction, and heart disease. Further, stroke, one of the leading risk factors for VCID, has a higher incidence in men than in women throughout much of the lifespan, though this trend is reversed at advanced ages. This review will highlight the need to take biological sex and common co-morbidities for VCID into account in both preclinical and clinical research. Given that there are currently no treatments available for VCID, it is critical that we understand how to mitigate risk factors for this devastating disease in both sexes.
Collapse
Affiliation(s)
- O J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - L S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - A J Custozzo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - K L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| |
Collapse
|
12
|
Selvaraj UM, Zuurbier KR, Whoolery CW, Plautz EJ, Chambliss KL, Kong X, Zhang S, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Mineo C, Shaul PW, Stowe AM. Selective Nonnuclear Estrogen Receptor Activation Decreases Stroke Severity and Promotes Functional Recovery in Female Mice. Endocrinology 2018; 159:3848-3859. [PMID: 30256928 PMCID: PMC6203892 DOI: 10.1210/en.2018-00600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Abstract
Estrogens provide neuroprotection in animal models of stroke, but uterotrophic effects and cancer risk limit translation. Classic estrogen receptors (ERs) serve as transcription factors, whereas nonnuclear ERs govern numerous cell processes and exert beneficial cardiometabolic effects without uterine or breast cancer growth in mice. Here, we determined how nonnuclear ER stimulation with pathway-preferential estrogen (PaPE)-1 affects stroke outcome in mice. Ovariectomized female mice received vehicle, estradiol (E2), or PaPE-1 before and after transient middle cerebral artery occlusion (tMCAo). Lesion severity was assessed with MRI, and poststroke motor function was evaluated through 2 weeks after tMCAo. Circulating, spleen, and brain leukocyte subpopulations were quantified 3 days after tMCAo by flow cytometry, and neurogenesis and angiogenesis were evaluated histologically 2 weeks after tMCAo. Compared with vehicle, E2 and PaPE-1 reduced infarct volumes at 3 days after tMCAo, though only PaPE-1 reduced leukocyte infiltration into the ischemic brain. Unlike E2, PaPE-1 had no uterotrophic effect. Both interventions had negligible effect on long-term poststroke neuronal or vascular plasticity. All mice displayed a decline in motor performance at 2 days after tMCAo, and vehicle-treated mice did not improve thereafter. In contrast, E2 and PaPE-1 treatment afforded functional recovery at 6 days after tMCAo and beyond. Thus, the selective activation of nonnuclear ER by PaPE-1 decreased stroke severity and improved functional recovery in mice without undesirable uterotrophic effects. The beneficial effects of PaPE-1 are also associated with attenuated neuroinflammation in the brain. PaPE-1 and similar molecules may warrant consideration as efficacious ER modulators providing neuroprotection without detrimental effects on the uterus or cancer risk.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kielen R Zuurbier
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cody W Whoolery
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik J Plautz
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiangmei Kong
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shanrong Zhang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neurology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
13
|
Moon GJ, Cho YH, Kim DH, Sung JH, Son JP, Kim S, Cha JM, Bang OY. Serum-mediated Activation of Bone Marrow-derived Mesenchymal Stem Cells in Ischemic Stroke Patients: A Novel Preconditioning Method. Cell Transplant 2018; 27:485-500. [PMID: 29774769 PMCID: PMC6038038 DOI: 10.1177/0963689718755404] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stroke induces complex and dynamic, local and systemic changes including inflammatory
reactions, immune responses, and repair and recovery processes. Mesenchymal stem cells
(MSCs) have been shown to enhance neurological recovery after stroke. We hypothesized that
serum factors play a critical role in the activation of bone marrow (BM) MSCs after stroke
such as by increasing proliferation, paracrine effects, and rejuvenation. Human MSCs
(hMSCs) were grown in fetal bovine serum (FBS), normal healthy control serum (NS), or
stroke patient serum (SS). MSCs cultured in growth medium with 10% SS or NS exhibited
higher proliferation indices than those cultured with FBS (P < 0.01).
FBS-, NS-, and SS-hMSCs showed differences in the expression of trophic factors; vascular
endothelial growth factor, glial cell–derived neurotrophic factor, and fibroblast growth
factor were densely expressed in samples cultured with SS (P < 0.01).
In addition, SS-MSCs revealed different cell cycle– or aging-associated messenger RNA
expression in a later passage, and β-galactosidase staining showed the senescence of MSCs
observed during culture expansion was lower in MSCs cultured with SS than those cultured
with NS or FBS (P < 0.01). Several proteins related to the activity of
receptors, growth factors, and cytokines were more prevalent in the serum of stroke
patients than in that of normal subjects. Neurogenesis and angiogenesis were markedly
increased in rats that had received SS-MSCs (P < 0.05), and these rats
showed significant behavioral improvements (P < 0.01). Our results
indicate that stroke induces a process of recovery via the activation of MSCs. Culture
methods for MSCs using SS obtained during the acute phase of a stroke could constitute a
novel MSC activation method that is feasible and efficient for the neurorestoration of
stroke.
Collapse
Affiliation(s)
- Gyeong Joon Moon
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,2 Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea.,3 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Buk-gu, Daegu, South Korea
| | - Yeon Hee Cho
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Dong Hee Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Ji Hee Sung
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jeong Pyo Son
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Sooyoon Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jae Min Cha
- 6 Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Oh Young Bang
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,7 Department of Neurology, Samsung Medical Center, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| |
Collapse
|
14
|
Tolahunase MR, Sagar R, Faiq M, Dada R. Yoga- and meditation-based lifestyle intervention increases neuroplasticity and reduces severity of major depressive disorder: A randomized controlled trial. Restor Neurol Neurosci 2018; 36:423-442. [PMID: 29614706 DOI: 10.3233/rnn-170810] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Madhuri R. Tolahunase
- Department of Anatomy, Lab for Molecular Reproduction and Genetics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rajesh Sagar
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Muneeb Faiq
- Department of Anatomy, Lab for Molecular Reproduction and Genetics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rima Dada
- Department of Anatomy, Lab for Molecular Reproduction and Genetics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
15
|
Sadaie MR, Farhoudi M, Zamanlu M, Aghamohammadzadeh N, Amouzegar A, Rosenbaum RE, Thomas GA. What does the research say about androgen use and cerebrovascular events? Ther Adv Drug Saf 2018; 9:439-455. [PMID: 30364888 DOI: 10.1177/2042098618773318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
Many studies have investigated the benefits of androgen therapy and neurosteroids in aging men, while concerns remain about the potential associations of exogenous steroids and incidents of cerebrovascular events and ischemic stroke (IS). Testosterone is neuroprotective, neurotrophic and a potent stimulator of neuroplasticity. These benefits are mediated primarily through conversion of a small amount of testosterone to estradiol by the catalytic activity of estrogen synthetase (aromatase cytochrome P450 enzyme). New studies suggest that abnormal serum levels of the nonaromatized potent metabolite of testosterone, either high or low dihydrotestosterone (DHT), is a risk factor for stroke. Associations between pharmacologic androgen use and the incidence of IS are questionable, because a significant portion of testosterone is converted to DHT. There is also insufficient evidence to reject a causal relationship between the pro-testosterone adrenal androgens and incidence of IS. Moreover, vascular intima-media thickness, which is a predictor of stroke and myocardial symptoms, has correlations with sex hormones. Current diagnostic and treatment criteria for androgen therapy for cerebrovascular complications are unclear. Confounding variables, including genetic and metabolic alterations of the key enzymes of steroidogenesis, ought to be considered. Information extracted from pharmacogenetic testing may aid in expounding the protective-destructive properties of neurosteroids, as well as the prognosis of androgen therapy, in particular their cerebrovascular outcomes. This investigative review article addresses relevant findings of the clinical and experimental investigations of androgen therapy, emphasizes the significance of genetic testing of androgen responsiveness towards individualized therapy in post-IS injuries as well as identifying pertinent questions.
Collapse
Affiliation(s)
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masumeh Zamanlu
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Aghamohammadzadeh
- Department of Endocrinology, Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atieh Amouzegar
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Gary A Thomas
- Penn State Hershey Neurology, Penn State University, PA, USA
| |
Collapse
|
16
|
Liang Y, Chen J, Zheng X, Chen Z, Liu Y, Li S, Fang X. Ultrasound-Mediated Kallidinogenase-Loaded Microbubble Targeted Therapy for Acute Cerebral Infarction. J Stroke Cerebrovasc Dis 2018; 27:686-696. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.09.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/24/2017] [Accepted: 09/29/2017] [Indexed: 10/18/2022] Open
|
17
|
|
18
|
Ponti G, Farinetti A, Marraudino M, Panzica G, Gotti S. Sex Steroids and Adult Neurogenesis in the Ventricular-Subventricular Zone. Front Endocrinol (Lausanne) 2018; 9:156. [PMID: 29686651 PMCID: PMC5900029 DOI: 10.3389/fendo.2018.00156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/22/2018] [Indexed: 12/28/2022] Open
Abstract
The forebrain ventricular-subventricular zone (V-SVZ) continuously generates new neurons throughout life. Neural stem cells (type B1 cells) along the lateral ventricle become activated, self-renew, and give rise to proliferating precursors which progress along the neurogenic lineage from intermediate progenitors (type C cells) to neuroblasts (type A cells). Neuroblasts proliferate and migrate into the olfactory bulb and differentiate into different interneuronal types. Multiple factors regulate each step of this process. Newly generated olfactory bulb interneurons are an important relay station in the olfactory circuits, controlling social recognition, reproductive behavior, and parental care. Those behaviors are strongly sexually dimorphic and changes throughout life from puberty through aging and in the reproductive age during estrous cycle and gestation. Despite the key role of sex hormones in regulating those behaviors, their contribution in modulating adult neurogenesis in V-SVZ is underestimated. Here, we compare the literature highlighting the sexual dimorphism and the differences across the physiological phases of the animal for the different cell types and steps through the neurogenic lineage.
Collapse
Affiliation(s)
- Giovanna Ponti
- Department of Veterinary Sciences, University of Turin, Grugliasco,Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- *Correspondence: Giovanna Ponti,
| | - Alice Farinetti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - GianCarlo Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, Turin, Italy
| |
Collapse
|
19
|
Akhoundzadeh K, Vakili A, Sameni HR, Vafaei AA, Rashidy-Pour A, Safari M, Mohammadkhani R. Effects of the combined treatment of bone marrow stromal cells with mild exercise and thyroid hormone on brain damage and apoptosis in a mouse focal cerebral ischemia model. Metab Brain Dis 2017; 32:1267-1277. [PMID: 28547077 DOI: 10.1007/s11011-017-0034-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 05/16/2017] [Indexed: 01/20/2023]
Abstract
This study examined whether post-stroke bone marrow stromal cells (BMSCs) therapy combined with exercise (EX) and/or thyroid hormone (TH) could reduce brain damage in an experimental ischemic stroke in mice. Focal cerebral ischemia was induced under Laser Doppler Flowmetry (LDF) guide by 45 min of middle cerebral artery occlusion (MCAO), followed by 7 days of reperfusion in albino mice. BMSCs were injected into the right cerebral ventricle 24 h after MCAO, followed by daily injection of T3 (20 μg/100 g weight S.C) and 6 days of running on a treadmill. Infarct size, neurobehavioral test, TUNEL and BrdU positive cells were evaluated at 7 days after MCAO. Treatment with BMSCs and mild EX alone significantly reduced the infarct volume by 23% and 44%, respectively (both, p < 0.001). The BMSCs + TH, BMSCs + EX, and BMSCs + EX + TH combination therapies significantly reduced the infarct volume by 26%, 51%, and 70%, respectively (all, p < 0.001). A significant improvement in the neurobehavioral functioning was observed in the EX, BMSCs + EX, and BMSCs + EX+ TH groups (p < 0.001). The number of TUNEL-positive cells (a marker of apoptosis) was significantly reduced in the EX, BMSCs, BMSCs + EX, BMSCs + TH, and BMSCs + EX + TH groups (all, p < 0.001). Moreover, the combination therapy considerably increased BrdU-labeled cells in the subventricular zone (SVZ) (p < 0.01). Our findings indicated that the combined treatment of BMSCs with mild EX and TH more efficiently reduces the cerebral infarct size after stroke. More likely, these effects mediate via enchaining generation of new neuronal cells and the attenuation of apoptosis in ischemia stroke in young mice.
Collapse
Affiliation(s)
- Kobar Akhoundzadeh
- Research Center and Department of Physiology, Faculty of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abedin Vakili
- Research Center and Department of Physiology, Faculty of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Hamid Reza Sameni
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Abbas Ali Vafaei
- Research Center and Department of Physiology, Faculty of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center and Department of Physiology, Faculty of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Manouchehr Safari
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Razieh Mohammadkhani
- Research Center and Department of Physiology, Faculty of Medicine, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
20
|
Jeton F, Soliz J, Marchant D, Joseph V, Richalet JP, Pichon A, Voituron N. Increased ventilation in female erythropoietin-deficient mouse line is not progesterone and estrous stage-dependent. Respir Physiol Neurobiol 2017; 245:98-104. [PMID: 28735074 DOI: 10.1016/j.resp.2017.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 01/21/2023]
Abstract
Previous studies suggest that chronic erythropoietin (Epo) deficiency in male mice does not alter normoxic/hypoxic ventilation. As effects of Epo are sex specific and as progesterone could be a respiratory stimulant, we evaluated the impact of Epo deficiency and its possible interaction with progesterone in ventilatory control in female mice during estrous cycle phases. Compared to wild type (WT) animals, Epo-TAgh female mice exhibited higher ventilation in hypoxia. However, when data were separated into luteal and follicular phases of the estrous cycle, basal ventilation and hypoxic ventilation were not different in both mice strains. As progesterone is known to be a potent respiratory stimulant, additional experiments were performed to elucidate its role. Interestingly, after mifepristone treatment, HVR was not modified in WT and Epo-TAgh mice, showing that the ventilatory stimulation observed in females was not directly mediated by progesterone. We conclude that Epo-TAgh female mice show no estrous stage-dependent increase of ventilatory control and progesterone independent response to hypoxia.
Collapse
Affiliation(s)
- Florine Jeton
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, EA 2363, 93017 Bobigny, France; Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, France
| | - Jorge Soliz
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Dominique Marchant
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, EA 2363, 93017 Bobigny, France
| | - Vincent Joseph
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Jean-Paul Richalet
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, EA 2363, 93017 Bobigny, France; Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, France
| | - Aurélien Pichon
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, EA 2363, 93017 Bobigny, France; Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, France
| | - Nicolas Voituron
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, EA 2363, 93017 Bobigny, France; Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, France.
| |
Collapse
|
21
|
Pino A, Fumagalli G, Bifari F, Decimo I. New neurons in adult brain: distribution, molecular mechanisms and therapies. Biochem Pharmacol 2017; 141:4-22. [PMID: 28690140 DOI: 10.1016/j.bcp.2017.07.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022]
Abstract
"Are new neurons added in the adult mammalian brain?" "Do neural stem cells activate following CNS diseases?" "How can we modulate their activation to promote recovery?" Recent findings in the field provide novel insights for addressing these questions from a new perspective. In this review, we will summarize the current knowledge about adult neurogenesis and neural stem cell niches in healthy and pathological conditions. We will first overview the milestones that have led to the discovery of the classical ventricular and hippocampal neural stem cell niches. In adult brain, new neurons originate from proliferating neural precursors located in the subventricular zone of the lateral ventricles and in the subgranular zone of the hippocampus. However, recent findings suggest that new neuronal cells can be added to the adult brain by direct differentiation (e.g., without cell proliferation) from either quiescent neural precursors or non-neuronal cells undergoing conversion or reprogramming to neuronal fate. Accordingly, in this review we will also address critical aspects of the newly described mechanisms of quiescence and direct conversion as well as the more canonical activation of the neurogenic niches and neuroblast reservoirs in pathological conditions. Finally, we will outline the critical elements involved in neural progenitor proliferation, neuroblast migration and differentiation and discuss their potential as targets for the development of novel therapeutic drugs for neurodegenerative diseases.
Collapse
Affiliation(s)
- Annachiara Pino
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Italy.
| |
Collapse
|
22
|
Pretreatment with minocycline restores neurogenesis in the subventricular zone and subgranular zone of the hippocampus after ketamine exposure in neonatal rats. Neuroscience 2017; 352:144-154. [DOI: 10.1016/j.neuroscience.2017.03.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/20/2017] [Accepted: 03/26/2017] [Indexed: 12/26/2022]
|
23
|
Abstract
The brain has long been known as a dimorphic organ and as a target of sex steroids. It is also a site for their synthesis. Sex steroids in numerous ways can modify cerebral physiology, and along with many processes adult neurogenesis is also modulated by sex steroids. This review will focus on the effects of the main steroids, estrogens, androgens and progestogens, and unveil some aspects of their partly disclosed mechanisms of actions. Gonadal steroids act on different steps of neurogenesis: cell proliferation seems to be increased by estrogens only, while androgens and progestogens favor neuronal renewal by increasing cell survival; differentiation is a common target. Aging is characterized by a cognitive deficiency, paralleled by a decrease in the rate of neuronal renewal and in the levels of circulating gonadal hormones. Therefore, the effects of gonadal hormones on the aging brain are important to consider. The review will also be expanded to related molecules which are agonists to the nuclear receptors. Sex steroids can modify adult neuronal renewal and the extensive knowledge of their actions on neurogenesis is essential, as it can be a leading pathway to therapeutic perspectives.
Collapse
Affiliation(s)
- Christine Heberden
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.
| |
Collapse
|
24
|
Stoop W, De Geyter D, Verachtert S, Brouwers S, Verdood P, De Keyser J, Kooijman R. Post-stroke treatment with 17β-estradiol exerts neuroprotective effects in both normotensive and hypertensive rats. Neuroscience 2017; 348:335-345. [DOI: 10.1016/j.neuroscience.2017.02.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/01/2022]
|
25
|
The choroid plexus as a sex hormone target: Functional implications. Front Neuroendocrinol 2017; 44:103-121. [PMID: 27998697 DOI: 10.1016/j.yfrne.2016.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/25/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022]
Abstract
The choroid plexuses (CPs) are highly vascularized branched structures that protrude into the ventricles of the brain, and form a unique interface between the blood and the cerebrospinal fluid (CSF). In recent years, novel functions have been attributed to this tissue such as in immune and chemical surveillance of the central nervous system, brain development, adult neurogenesis and circadian rhythm regulation. Sex hormones (SH) are widely recognized as modulators in several neurodegenerative diseases, and there is evidence that estrogens and androgens regulate several fundamental biological functions in the CPs. Therefore, SH are likely to affect the composition of the CSF impacting on brain homeostasis. This review will look at implications of the CPs' sex-related specificities.
Collapse
|
26
|
Martín-Aragón Baudel MAS, Poole AV, Darlison MG. Chloride co-transporters as possible therapeutic targets for stroke. J Neurochem 2016; 140:195-209. [PMID: 27861901 DOI: 10.1111/jnc.13901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of death and disability worldwide. The major type of stroke is an ischaemic one, which is caused by a blockage that interrupts blood flow to the brain. There are currently very few pharmacological strategies to reduce the damage and social burden triggered by this pathology. The harm caused by the interruption of blood flow to the brain unfolds in the subsequent hours and days, so it is critical to identify new therapeutic targets that could reduce neuronal death associated with the spread of the damage. Here, we review some of the key molecular mechanisms involved in the progression of neuronal death, focusing on some new and promising studies. In particular, we focus on the potential of the chloride co-transporter (CCC) family of proteins, mediators of the GABAergic response, both during the early and later stages of stroke, to promote neuroprotection and recovery. Different studies of CCCs during the chronic and recovery phases post-stroke reveal the importance of timing when considering CCCs as potential neuroprotective and/or neuromodulator targets. The molecular regulatory mechanisms of the two main neuronal CCCs, NKCC1 and KCC2, are further discussed as an indirect approach for promoting neuroprotection and neurorehabilitation following an ischaemic insult. Finally, we mention the likely importance of combining different strategies in order to achieve more effective therapies.
Collapse
Affiliation(s)
| | - Amy V Poole
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| | - Mark G Darlison
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| |
Collapse
|
27
|
Guan J, Zhang S, Zhou Q, Yuan Z, Lu Z. Effect of thrombin preconditioning on migration of subventricular zone-derived cells after intracerebral hemorrhage in rats. Neurol Res 2016; 38:809-16. [PMID: 27477964 DOI: 10.1080/01616412.2016.1210356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To investigate the effect of thrombin preconditioning (TPC) on the intracerebral hemorrhage (ICH)-induced proliferation, migration, and function of subventriclular zone (SVZ) cells and to find new strategies that enhance endogenous neurogenesis after ICH. METHODS Male Sprague-Dawley rats were randomly divided into 3 groups (ICH, TPC, and control group). Rats of each group were randomly divided into 5 subgroups (3-d, 7-d, 14-d, 21-d, and 28-d subgroup). ICH was caused by intrastrial stereotactic administration of collagenase type IV. Brdu was used to label newborn SVZ cells. Organotypic brain slices were cultured to dynamically observe the migration of SVZ cells at living brain tissue. Migration of Dil-labeled SVZ cells in living brain slices was traced by time-lapse microscopy. To assess whether SVZ cells migrating to injured striatum had the ability to form synapses with other cells, brain slices from each group were double immunolabeled with Brdu and synapsin I. RESULTS The number of Brdu-positive cells markedly increased in the ipsilateral SVZ and striatum 3 days after TPC, peaked at 14 days (P < 0.01), continued to 21 days, and then gradually decreased at 28 days with significant difference compared to the ICH group at each time point (P < 0.01). Migration of Dil-labeled SVZ cells in brain slices in each group was observed and imaged during a 12-h period. Dil-labeled SVZ cells in the TPC group were observed to migrate laterally toward striatum with time with a faster velocity compared to the ICH group (P < 0.01). Our study also demonstrated that TPC induced strong colocalization of Brdu and synapsin I in the ipsilateral striatum between 3 and 28 days after injury.TPC made colocalization of Brdu and synapsin I appear earlier and continue for a longer time compared to the ICH group. CONCLUSIONS Our results demonstrated that TPC could promote proliferation, migration, and function of SVZ cells after ICH, which may provide a new idea for enhancing endogenous neurogenesis and developing new therapeutic strategies against ICH-induced brain injury.
Collapse
Affiliation(s)
- Jingxia Guan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Shaofeng Zhang
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Qin Zhou
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zhenhua Yuan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zuneng Lu
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
28
|
He Z, Cui L, Paule MG, Ferguson SA. Estrogen Selectively Mobilizes Neural Stem Cells in the Third Ventricle Stem Cell Niche of Postnatal Day 21 Rats. Mol Neurobiol 2016; 52:927-33. [PMID: 26041664 DOI: 10.1007/s12035-015-9244-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neuroprotective properties of stem cells have been described for various pathophysiological states. Here, we determined the effects of exogenous perinatal estrogen treatment on endogenous neural stem cell activity in the third ventricle stem cell niche (3VSCN) and the caudal third ventricle (C3V). Pregnant Sprague-Dawley rats were gavaged with ethinyl estradiol (EE2, 10 μg/kg/day) or vehicle on gestational days 6-21, and their offspring were similarly treated from birth to weaning on postnatal day 21. At weaning, neural stem cell activity was investigated using the stem cell markers nestin, Ki-67, phosphohistone H3 (PHH3), and doublecortin (DCX). The 3VSCN was characterized by nestin labeling, but little DCX labeling, while both the subventricular (SVZ) and subgranular zones (SGZ) displayed robust DCX expression. Ki-67 cell counts in the 3VSCN were 2.2 to 6.4 times those of the C3V. In the 3VSCN, EE2 treatment significantly increased Ki-67, PHH3, and co-labeled cell counts by 135-207 %, effects which appeared stronger in females. EE2 treatment had only marginally significant effects in the C3V, mildly increasing PHH3 and co-labeled cell counts. Perinatal estrogen treatment selectively increased and mobilized proliferative cells in the 3VSCN at weaning, potentially providing increased neuroprotection. Because PHH3 cells are thought to be in the mitotic phase of the cell cycle and Ki-67 cells can be found in most phases of the cycle, the effect of estrogen treatment on 3VSCN cells appears to involve enhancement of mitosis.
Collapse
Affiliation(s)
- Zhen He
- HFT-132, Division of Neurotoxicology, National Center for Toxicological Research/FDA, 3900 NCTR Road, Jefferson, AR, 72079, USA,
| | | | | | | |
Collapse
|
29
|
Carpenter RS, Iwuchukwu I, Hinkson CL, Reitz S, Lee W, Kukino A, Zhang A, Pike MM, Ardelt AA. High-dose estrogen treatment at reperfusion reduces lesion volume and accelerates recovery of sensorimotor function after experimental ischemic stroke. Brain Res 2016; 1639:200-13. [PMID: 26995494 DOI: 10.1016/j.brainres.2016.01.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/03/2015] [Accepted: 01/17/2016] [Indexed: 01/15/2023]
Abstract
Estrogens have previously been shown to protect the brain against acute ischemic insults, by potentially augmenting cerebrovascular function after ischemic stroke. The current study hypothesized that treatment with sustained release of high-dose 17β-estradiol (E2) at the time of reperfusion from middle cerebral artery occlusion (MCAO) in rats would attenuate reperfusion injury, augment post-stroke angiogenesis and cerebral blood flow, and attenuate lesion volume. Female Wistar rats underwent ovariectomy, followed two weeks later by transient, two-hour right MCAO (tMCAO) and treatment with E2 (n=13) or placebo (P; n=12) pellets starting at reperfusion. E2 treatment resulted in significantly smaller total lesion volume, smaller lesions within striatal and cortical brain regions, and less atrophy of the ipsilateral hemisphere after six weeks of recovery. E2-treated animals exhibited accelerated recovery of contralateral forelimb sensorimotor function in the cylinder test. Magnetic resonance imaging (MRI) showed that E2 treatment reduced the formation of lesion cysts, decreased lesion volume, and increased lesional cerebral blood flow (CBF). K(trans), a measure of vascular permeability, was increased in the lesions. This finding, which represents lesion neovascularization, was not altered by E2 treatment. Ischemic stroke-related angiogenesis and vessel formation was confirmed with immunolabeling of brain tissue and was not altered with E2 treatment. In summary, E2 treatment administered immediately following reperfusion significantly reduced lesion size, cyst formation, and brain atrophy while improving lesional CBF and accelerating recovery of functional deficits in a rat model of ischemic stroke.
Collapse
Affiliation(s)
| | | | | | - Sydney Reitz
- The College, University of Chicago, Chicago, IL, USA
| | - Wonhee Lee
- The College, University of Chicago, Chicago, IL, USA
| | - Ayaka Kukino
- Advanced Imaging Research Center, Oregon Health Sciences University, Portland, OR, USA
| | - An Zhang
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health Sciences University, Portland, OR, USA
| | | |
Collapse
|
30
|
Lindvall O, Kokaia Z. Neurogenesis following Stroke Affecting the Adult Brain. Cold Spring Harb Perspect Biol 2015; 7:7/11/a019034. [PMID: 26525150 DOI: 10.1101/cshperspect.a019034] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A bulk of experimental evidence supports the idea that the stroke-damaged adult brain makes an attempt to repair itself by producing new neurons also in areas where neurogenesis does not normally occur (e.g., the striatum and cerebral cortex). Knowledge about mechanisms regulating the different steps of neurogenesis after stroke is rapidly increasing but still incomplete. The functional consequences of stroke-induced neurogenesis and the level of integration of the new neurons into existing neural circuitries are poorly understood. To have a substantial impact on the recovery after stroke, this potential mechanism for self-repair needs to be enhanced, primarily by increasing the survival and differentiation of the generated neuroblasts. Moreover, for efficient repair, optimization of neurogenesis most likely needs to be combined with promotion of other endogenous neuroregenerative responses (e.g., protection and sprouting of remaining mature neurons, transplantation of neural stem/progenitor cells [NSPC]-derived neurons and glia cells, and modulation of inflammation).
Collapse
Affiliation(s)
- Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden
| |
Collapse
|
31
|
Tan S, Zhi P, Luo Z, Shi J. Severe instead of mild hyperglycemia inhibits neurogenesis in the subventricular zone of adult rats after transient focal cerebral ischemia. Neuroscience 2015; 303:138-48. [DOI: 10.1016/j.neuroscience.2015.06.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 01/04/2023]
|
32
|
Khan MM, Wakade C, de Sevilla L, Brann DW. Selective estrogen receptor modulators (SERMs) enhance neurogenesis and spine density following focal cerebral ischemia. J Steroid Biochem Mol Biol 2015; 146:38-47. [PMID: 24815952 PMCID: PMC4419701 DOI: 10.1016/j.jsbmb.2014.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/23/2014] [Accepted: 05/02/2014] [Indexed: 12/23/2022]
Abstract
Selective estrogen receptor modulators (SERMs) have been reported to enhance synaptic plasticity and improve cognitive performance in adult rats. SERMs have also been shown to induce neuroprotection against cerebral ischemia and other CNS insults. In this study, we sought to determine whether acute regulation of neurogenesis and spine remodeling could be a novel mechanism associated with neuroprotection induced by SERMs following cerebral ischemia. Toward this end, ovariectomized adult female rats were either implanted with pellets of 17β-estradiol (estrogen) or tamoxifen, or injected with raloxifene. After one week, cerebral ischemia was induced by the transient middle-cerebral artery occlusion (MCAO) method. Bromodeoxyuridine (BrdU) was injected to label dividing cells in brain. We analyzed neurogenesis and spine density at day-1 and day-5 post MCAO. In agreement with earlier findings, we observed a robust induction of neurogenesis in the ipsilateral subventricular zone (SVZ) of both the intact as well as ovariectomized female rats following MCAO. Interestingly, neurogenesis in the ipsilateral SVZ following ischemia was significantly higher in estrogen and raloxifene-treated animals compared to placebo-treated rats. In contrast, this enhancing effect on neurogenesis was not observed in tamoxifen-treated rats. Finally, both SERMs, as well as estrogen significantly reversed the spine density loss observed in the ischemic cortex at day-5 post ischemia. Taken, together these results reveal a profound structural remodeling potential of SERMs in the brain following cerebral ischemia. This article is part of a Special Issue entitled "Sex steroids and brain disorders".
Collapse
Affiliation(s)
- Mohammad M Khan
- Department of Biochemistry, Faculty of Medicine, Zaiwa University, AZ-Zawia, Libya
| | - Chandramohan Wakade
- Department of Physical Therapy, Georgia Regents University, Augusta, GA 30912, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Liesl de Sevilla
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
33
|
Hansberg-Pastor V, González-Arenas A, Piña-Medina AG, Camacho-Arroyo I. Sex Hormones Regulate Cytoskeletal Proteins Involved in Brain Plasticity. Front Psychiatry 2015; 6:165. [PMID: 26635640 PMCID: PMC4653291 DOI: 10.3389/fpsyt.2015.00165] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/02/2015] [Indexed: 01/22/2023] Open
Abstract
In the brain of female mammals, including humans, a number of physiological and behavioral changes occur as a result of sex hormone exposure. Estradiol and progesterone regulate several brain functions, including learning and memory. Sex hormones contribute to shape the central nervous system by modulating the formation and turnover of the interconnections between neurons as well as controlling the function of glial cells. The dynamics of neuron and glial cells morphology depends on the cytoskeleton and its associated proteins. Cytoskeletal proteins are necessary to form neuronal dendrites and dendritic spines, as well as to regulate the diverse functions in astrocytes. The expression pattern of proteins, such as actin, microtubule-associated protein 2, Tau, and glial fibrillary acidic protein, changes in a tissue-specific manner in the brain, particularly when variations in sex hormone levels occur during the estrous or menstrual cycles or pregnancy. Here, we review the changes in structure and organization of neurons and glial cells that require the participation of cytoskeletal proteins whose expression and activity are regulated by estradiol and progesterone.
Collapse
Affiliation(s)
- Valeria Hansberg-Pastor
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Ana Gabriela Piña-Medina
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México , Mexico City , Mexico
| |
Collapse
|
34
|
Mirzamohammadi S, Aali E, Najafi R, Kamarul T, Mehrabani M, Aminzadeh A, Sharifi AM. Effect of 17β-estradiol on mediators involved in mesenchymal stromal cell trafficking in cell therapy of diabetes. Cytotherapy 2014; 17:46-57. [PMID: 25457279 DOI: 10.1016/j.jcyt.2014.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have shown great promise for cell therapy of a wide range of diseases such as diabetes. However, insufficient viability of transplanted cells reaching to damaged tissues has limited their potential therapeutic effects. Expression of estrogen receptors on stem cells may suggest a role for 17β-estradiol (E2) in regulating some functions in these cells. There is evidence that E2 enhances homing of stem cells. Induction of hypoxia-inducible factor-1α (HIF-1α) by E2 and the profound effect of HIF-1α on migration of cells have previously been demonstrated. We investigated the effect of E2 on major mediators involved in trafficking and subsequent homing of MSCs both in vitro and in vivo in diabetic rats. METHODS E2 has been selected to improve the poor migration capacity of MSCs toward sites of injury. MSCs were incubated with different concentrations of E2 for varying periods of time to investigate whether estradiol treatment could be effective to enhance the efficiency of MSC transplantation. RESULTS E2 significantly enhanced the viability of the cells that were blocked by ICI 182,780 (estrogen receptor antagonist). E2 also increased HIF-1α, CXC chemokine receptor 4 and C-C chemokine receptor 2 protein and messenger RNA levels measured by Western blot and reverse transcription-polymerase chain reaction. The enzymatic activity of matrix metalloproteinase 2 and metalloproteinase 9 was elevated in E2-treated cells through the use of gelatin zymography. Finally, the improved migration capacity of E2-treated MSCs was evaluated with the use of a Boyden chamber and in vivo migration assays. CONCLUSIONS Our data support that conditioning of MSCs with E2 promotes migration of cells in cultured MSCs in vitro and in a diabetic rat model in vivo through regulation of major mediators of cell trafficking.
Collapse
Affiliation(s)
- Solmaz Mirzamohammadi
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Aali
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Najafi
- Department of Molecular Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Tunku Kamarul
- Tissue Engineering Group (TEG) and Research, National Orthopedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopedics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mehrnaz Mehrabani
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Aminzadeh
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Sharifi
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Cell Therapy, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Lei S, Zhang P, Li W, Gao M, He X, Zheng J, Li X, Wang X, Wang N, Zhang J, Qi C, Lu H, Chen X, Liu Y. Pre- and posttreatment with edaravone protects CA1 hippocampus and enhances neurogenesis in the subgranular zone of dentate gyrus after transient global cerebral ischemia in rats. ASN Neuro 2014; 6:6/6/1759091414558417. [PMID: 25388889 PMCID: PMC4357607 DOI: 10.1177/1759091414558417] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Edaravone is clinically used for treatment of patients with acute cerebral infarction. However, the effect of double application of edaravone on neurogenesis in the hippocampus following ischemia remains unknown. In the present study, we explored whether pre- and posttreatment of edaravone had any effect on neural stem/progenitor cells (NSPCs) in the subgranular zone of hippocampus in a rat model of transient global cerebral ischemia and elucidated the potential mechanism of its effects. Male Sprague-Dawley rats were divided into three groups: sham-operated (n = 15), control (n = 15), and edaravone-treated (n = 15) groups. Newly generated cells were labeled by 5-bromo-2-deoxyuridine. Immunohistochemistry was used to detect neurogenesis. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling was used to detect cell apoptosis. Reactive oxygen species (ROS) were detected by 2,7-dichlorofluorescien diacetate assay in NSPCs in vitro. Hypoxia-inducible factor-1α (HIF-1α) and cleaved caspase-3 proteins were quantified by western blot analysis. Treatment with edaravone significantly increased the number of NSPCs and newly generated neurons in the subgranular zone (p < .05). Treatment with edaravone also decreased apoptosis of NSPCs (p < .01). Furthermore, treatment with edaravone significantly decreased ROS generation and inhibited HIF-1α and cleaved caspase-3 protein expressions. These findings indicate that pre- and posttreatment with edaravone enhances neurogenesis by protecting NSPCs from apoptosis in the hippocampus, which is probably mediated by decreasing ROS generation and inhibiting protein expressions of HIF-1α and cleaved caspase-3 after cerebral ischemia.
Collapse
Affiliation(s)
- Shan Lei
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Weisong Li
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Ming Gao
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Juan Zheng
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xu Li
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xiao Wang
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Ning Wang
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Junfeng Zhang
- Department of Anatomy, Xi'an Medical University, Xi'an, China
| | - Cunfang Qi
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University School of Medicine, Xi'an, China
| |
Collapse
|
36
|
Merson TD, Bourne JA. Endogenous neurogenesis following ischaemic brain injury: insights for therapeutic strategies. Int J Biochem Cell Biol 2014; 56:4-19. [PMID: 25128862 DOI: 10.1016/j.biocel.2014.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/18/2014] [Accepted: 08/04/2014] [Indexed: 01/19/2023]
Abstract
Ischaemic stroke is among the most common yet most intractable types of central nervous system (CNS) injury in the adult human population. In the acute stages of disease, neurons in the ischaemic lesion rapidly die and other neuronal populations in the ischaemic penumbra are vulnerable to secondary injury. Multiple parallel approaches are being investigated to develop neuroprotective, reparative and regenerative strategies for the treatment of stroke. Accumulating evidence indicates that cerebral ischaemia initiates an endogenous regenerative response within the adult brain that potentiates adult neurogenesis from populations of neural stem and progenitor cells. A major research focus has been to understand the cellular and molecular mechanisms that underlie the potentiation of adult neurogenesis and to appreciate how interventions designed to modulate these processes could enhance neural regeneration in the post-ischaemic brain. In this review, we highlight recent advances over the last 5 years that help unravel the cellular and molecular mechanisms that potentiate endogenous neurogenesis following cerebral ischaemia and are dissecting the functional importance of this regenerative mechanism following brain injury. This article is part of a Directed Issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
Affiliation(s)
- Tobias D Merson
- Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, 30 Royal Parade, Parkville, VIC 3010, Australia.
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, Building 75, Level 1 North STRIP 1, Clayton, VIC 3800, Australia.
| |
Collapse
|
37
|
Ström JO, Ingberg E. Impact of methodology on estrogens' effects on cerebral ischemia in rats: an updated meta-analysis. BMC Neurosci 2014; 15:22. [PMID: 24495535 PMCID: PMC3975994 DOI: 10.1186/1471-2202-15-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/29/2014] [Indexed: 12/15/2022] Open
Abstract
Background Although most animal stroke studies have demonstrated potent neuroprotective effects of estrogens, there are a number of articles reporting the opposite. In 2009, we made the case that this dichotomy was related to administered estrogen dose. Several other suggestions for the discordant results have also been propagated, including the age of the experimental animals and the length of hypoestrogenicity prior to estrogen administration. These two suggestions have gained much popularity, probably because of their kinship with the window of opportunity hypothesis, which is commonly used to explain the analogous dichotomy among human studies. We were therefore encouraged to perform an updated meta-analysis, and to improve it by including all relevant variables in a large multiple regression model, where the impact of confounders could be controlled for. Results The multiple regression model revealed an indisputable impact of estrogen administration mode on the effects of estrogens in ischemic stroke. Subcutaneous slow-release pellets differed from the injection and silastic capsule treatments in terms of impact of estrogens on ischemic stroke, showing that the first mentioned were more prone to render estrogens damaging. Neither the use of elderly animals nor the adoption of longer wash-out periods influenced estrogens’ effects on experimental ischemic stroke in rats. Conclusions We conclude that the discordant results regarding estrogens’ effects in rat models of ischemic stroke are a consequence of differences in estrogen administration modes. These results are not only of importance for the ongoing debate regarding menopausal hormone therapy, but also have an important bearing on experimental stroke methodology and the apparent translational roadblock for suggested stroke interventions.
Collapse
Affiliation(s)
- Jakob O Ström
- Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro SE-703 62, Sweden.
| | | |
Collapse
|