1
|
Gaston-Breton R, Disdier C, Hagberg H, Mabondzo A. Hypoxia-ischemia and sexual dimorphism: modeling mitochondrial dysfunction using brain organoids. Cell Biosci 2025; 15:67. [PMID: 40413513 PMCID: PMC12103005 DOI: 10.1186/s13578-025-01402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/27/2025] [Indexed: 05/27/2025] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a leading cause of neurodevelopmental morbidities in full-term infants. There is strong evidence of sexual differences in hypoxic-ischemic (HI) injury where male neonates are at higher risk as they are subject to more pronounced neurological deficits and death than females. The cellular and molecular mechanisms underlying these sexual discrepancies in HI injury are poorly understood. Mitochondrial dysregulation has been increasingly explored in brain diseases and represents a major target during HI events. In this review, we discuss (1) different mitochondrial functions in the central nervous system (2), mitochondrial dysregulation in the context of HI injury (3), sex-dependent mitochondrial pathways in HIE and (4) modeling of mitochondrial dysfunction using human brain organoids. Gaining insight into these novel aspects of mitochondrial function will offer valuable understanding of brain development and neurological disorders such as HI injury, paving the way for the discovery and creation of new treatment approaches.
Collapse
Affiliation(s)
- Romane Gaston-Breton
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | - Clémence Disdier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | | | - Aloïse Mabondzo
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France.
| |
Collapse
|
2
|
Bahramiazar P, Abdollahzade N, Tartibian B, Ahmadiasl N, Yaghoob Nezhad F. The Role of Estrogen in Brain MicroRNAs Regulation. Adv Pharm Bull 2024; 14:819-835. [PMID: 40190672 PMCID: PMC11970499 DOI: 10.34172/apb.39216] [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: 08/28/2022] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose This review aims to elucidate the role of estrogen-sensitive microRNAs (miRNAs) in modulating brain functions and disorders, highlighting the protective effects of estrogen on the central nervous system. Methods A comprehensive literature review was conducted, examining the relationship between estrogen, miRNAs, and cognitive health. The study focused on experimental data comparing cognitive impairments between genders and the mechanisms of estrogen's effects on brain function. Results Cognitive impairments are less prevalent in women of reproductive age compared to men, indicating estrogen's neuroprotective role. Estrogen modulates gene expression through specific receptors, while miRNAs regulate approximately 30% of protein-coding genes in mammals. These miRNAs play critical roles in synaptic plasticity and neuronal survival. The review identifies several estrogen-sensitive miRNAs and their potential involvement in brain disorders. Conclusion The interplay between estrogen and miRNAs offers valuable insights into the molecular mechanisms underlying cognitive health and disease. Understanding these relationships may lead to novel therapeutic strategies for addressing various brain disorders, particularly those associated with hormonal changes and aging.
Collapse
Affiliation(s)
- Peyvand Bahramiazar
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naseh Abdollahzade
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Naser Ahmadiasl
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
3
|
Lear BA, Zhou KQ, Dhillon SK, Lear CA, Bennet L, Gunn AJ. Preventive, rescue and reparative neuroprotective strategies for the fetus and neonate. Semin Fetal Neonatal Med 2024; 29:101542. [PMID: 39472238 DOI: 10.1016/j.siny.2024.101542] [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: 12/13/2024]
Abstract
Neonatal encephalopathy remains a major contributor to death and disability around the world. Acute hypoxia-ischaemia before, during or after birth creates a series of events that can lead to neonatal brain injury. Understanding the evolution of injury underpinned the development of therapeutic hypothermia. This review discusses the determinants of injury, including maturity, the pattern of exposure to HI, impaired placental function, often associated with fetal growth restriction and in the long-term, socio-economic deprivation. Chorioamnionitis has been associated with the presence of NE, but it is important to note that experimentally, inflammation can either sensitize to greater neural injury after HI or alleviate injury, depending on its precise timing. As fetal surveillance tools improve it is likely that improved detection of specific pathways will offer future opportunities for preventive and reparative interventions in utero and after birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Kelly Q Zhou
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
4
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4 -associated aging phenotypes with the longevity-promoting intervention 17α-estradiol in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584678. [PMID: 38559059 PMCID: PMC10980056 DOI: 10.1101/2024.03.12.584678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The apolipoprotein ε4 allele ( APOE4 ) is associated with decreased longevity, increased vulnerability to age-related declines, and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4 -associated aging phenotypes. Here we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against age-related impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared for indices of aging phenotypes body-wide. Across peripheral and neural measures, APOE4 was associated with poorer outcomes. Notably, 17αE2 treatment improved outcomes in a genotype-dependent manner favoring APOE4 mice. These data demonstrate a positive APOE4 bias in 17αE2-mediated healthspan actions, suggesting that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with APOE4 genotype.
Collapse
|
5
|
Sunny A, James RR, Menon SR, Rayaroth S, Daniel A, Thompson NA, Tharakan B. Matrix Metalloproteinase-9 inhibitors as therapeutic drugs for traumatic brain injury. Neurochem Int 2024; 172:105642. [PMID: 38008261 DOI: 10.1016/j.neuint.2023.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality among young adults and the elderly. In the United States, TBI is responsible for around 30 percent of all injuries brought on by injuries in general. Vasogenic cerebral edema due to blood-brain barrier (BBB) dysfunction and the associated elevation of intracranial pressure (ICP) are some of the major causes of secondary injuries following traumatic brain injury. Matrix metalloproteinase-9 (MMP-9) is a therapeutic target for being an enzyme that degrades the proteins that make up a part of the microvascular basal lamina as well as inter-endothelial tight junctions of the blood-brain barrier. MMP-9-mediated BBB dysfunctions and the compromise of the BBB is a major pathway that leads the development of vasogenic cerebral edema, elevation of ICP, poor cerebral perfusion and brain herniation following traumatic brain injury. That makes MMP-9 an effective therapeutic target and endogenous or exogenous MMP-9 inhibitors as therapeutic drugs for preventing secondary brain damage after traumatic brain injury. Although our understanding of the mechanisms that underlie the primary and secondary stages of damage following a TBI has significantly improved in recent years, such information has not yet resulted in the successful development of novel pharmacological treatment options for traumatic brain injury. Recent pre-clinical and/or clinical studies have demonstrated that there are several compounds with specific or non-specific MMP-9 inhibitory properties either directly binding and inhibiting MMP-9 or by indirectly inhibiting MMP-9, with potential as therapeutic agents for traumatic brain injury. This article reviews the efficacy of several such medications and potential agents that include endogenous and exogeneous compounds that are at various levels of research and development. MMP-9-based therapeutic drug development has enormous potential in the pharmacological treatment of cerebral edema and/or neuronal injury resulting from traumatic brain injury.
Collapse
Affiliation(s)
- Angel Sunny
- Icahn School of Medicine at Mount Sinai, Elmhurst, NY, USA
| | | | | | | | - Abhijith Daniel
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Namita Ann Thompson
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
Gross KS, Lincoln CM, Anderson MM, Geiger GE, Frick KM. Extracellular matrix metalloproteinase-9 (MMP-9) is required in female mice for 17β-estradiol enhancement of hippocampal memory consolidation. Psychoneuroendocrinology 2022; 141:105773. [PMID: 35490640 PMCID: PMC9173600 DOI: 10.1016/j.psyneuen.2022.105773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/29/2022]
Abstract
Hippocampal plasticity and memory are modulated by the potent estrogen 17β-estradiol (E2). Research on the molecular mechanisms of hippocampal E2 signaling has uncovered multiple intracellular pathways that contribute to these effects, but few have questioned the role that extracellular signaling processes may play in E2 action. Modification of the extracellular matrix (ECM) by proteases like matrix metalloproteinase-9 (MMP-9) is critical for activity-dependent remodeling of synapses, and MMP-9 activity is required for hippocampal learning and memory. Yet little is known about the extent to which E2 regulates MMP-9 in the hippocampus, and the influence this interaction may have on hippocampal memory. Here, we examined the effects of hippocampal MMP-9 activity on E2-induced enhancement of spatial and object recognition memory consolidation. Post-training bilateral infusion of an MMP-9 inhibitor into the dorsal hippocampus of ovariectomized female mice blocked the enhancing effects of E2 on object placement and object recognition memory, supporting a role for MMP-9 in estrogenic regulation of memory consolidation. E2 also rapidly increased the activity of dorsal hippocampal MMP-9 without influencing its protein expression, providing further insight into hippocampal E2/MMP-9 interactions. Together, these results provide the first evidence that E2 regulates MMP-9 to modulate hippocampal memory and highlight the need to further study estrogenic regulation of extracellular modification.
Collapse
Affiliation(s)
| | | | | | | | - Karyn M. Frick
- Correspondence to: Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI 53211, USA. (K.M. Frick)
| |
Collapse
|
7
|
Tang T, Hu L, Liu Y, Fu X, Li J, Yan F, Cao S, Chen G. Sex-Associated Differences in Neurovascular Dysfunction During Ischemic Stroke. Front Mol Neurosci 2022; 15:860959. [PMID: 35431804 PMCID: PMC9012443 DOI: 10.3389/fnmol.2022.860959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/28/2022] [Indexed: 12/28/2022] Open
Abstract
Neurovascular units (NVUs) are basic functional units in the central nervous system and include neurons, astrocytes and vascular compartments. Ischemic stroke triggers not only neuronal damage, but also dissonance of intercellular crosstalk within the NVU. Stroke is sexually dimorphic, but the sex-associated differences involved in stroke-induced neurovascular dysfunction are studied in a limited extend. Preclinical studies have found that in rodent models of stroke, females have less neuronal loss, stronger repairing potential of astrocytes and more stable vascular conjunction; these properties are highly related to the cerebroprotective effects of female hormones. However, in humans, these research findings may be applicable only to premenopausal stroke patients. Women who have had a stroke usually have poorer outcomes compared to men, and because stoke is age-related, hormone replacement therapy for postmenopausal women may exacerbate stroke symptoms, which contradicts the findings of most preclinical studies. This stark contrast between clinical and laboratory findings suggests that understanding of neurovascular differences between the sexes is limited. Actually, apart from gonadal hormones, differences in neuroinflammation as well as genetics and epigenetics promote the sexual dimorphism of NVU functions. In this review, we summarize the confirmed sex-associated differences in NVUs during ischemic stroke and the possible contributing mechanisms. We also describe the gap between clinical and preclinical studies in terms of sexual dimorphism.
Collapse
Affiliation(s)
- Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Libin Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Liu
- Department of Ultrasonography, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Shenglong Cao,
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Gao Chen,
| |
Collapse
|
8
|
Chelluboina B, Chokkalla AK, Mehta SL, Morris-Blanco KC, Bathula S, Sankar S, Park JS, Vemuganti R. Tenascin-C induction exacerbates post-stroke brain damage. J Cereb Blood Flow Metab 2022; 42:253-263. [PMID: 34689646 PMCID: PMC9122520 DOI: 10.1177/0271678x211056392] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The role of tenascin-C (TNC) in ischemic stroke pathology is not known despite its prognostic association with cerebrovascular diseases. Here, we investigated the effect of TNC knockdown on post-stroke brain damage and its putative mechanism of action in adult mice of both sexes. Male and female C57BL/6 mice were subjected to transient middle cerebral artery occlusion and injected (i.v.) with either TNC siRNA or a negative (non-targeting) siRNA at 5 min after reperfusion. Motor function (beam walk and rotarod tests) was assessed between days 1 and 14 of reperfusion. Infarct volume (T2-MRI), BBB damage (T1-MRI with contrast), and inflammatory markers were measured at 3 days of reperfusion. The TNC siRNA treated cohort showed significantly curtailed post-stroke TNC protein expression, motor dysfunction, infarction, BBB damage, and inflammation compared to the sex-matched negative siRNA treated cohort. These results demonstrate that the induction of TNC during the acute period after stroke might be a mediator of post-ischemic inflammation and secondary brain damage independent of sex.
Collapse
Affiliation(s)
- Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | | | | - Sneha Sankar
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Jin Soo Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.,William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
9
|
Paul V, Krishnakumar S, Gowd GS, Nair SV, Koyakutty M, Paul-Prasanth B. Sex-Dependent Bioaccumulation of Nano Zinc Oxide and Its Adverse Effects on Sexual Behavior and Reproduction in Japanese Medaka. ACS APPLIED BIO MATERIALS 2021; 4:7408-7421. [DOI: 10.1021/acsabm.1c00575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Vinod Paul
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | | | | | - Shantikumar V. Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Manzoor Koyakutty
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Bindhu Paul-Prasanth
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| |
Collapse
|
10
|
Amirkhosravi L, Khaksari M, Sheibani V, Shahrokhi N, Ebrahimi MN, Amiresmaili S, Salmani N. Improved spatial memory, neurobehavioral outcomes, and neuroprotective effect after progesterone administration in ovariectomized rats with traumatic brain injury: Role of RU486 progesterone receptor antagonist. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:349-359. [PMID: 33995946 PMCID: PMC8087858 DOI: 10.22038/ijbms.2021.50973.11591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The contribution of classic progesterone receptors (PR) in interceding the neuroprotective efficacy of progesterone (P4) on the prevention of brain edema and long-time behavioral disturbances was assessed in traumatic brain injury (TBI). MATERIALS AND METHODS Female Wistar rats were ovariectomized and apportioned into 6 groups: sham, TBI, oil, P4, vehicle, and RU486. P4 or oil was injected following TBI. The antagonist of PR (RU486) or DMSO was administered before TBI. The brain edema and destruction of the blood-brain barrier (BBB) were determined. Intracranial pressure (ICP), cerebral perfusion pressure (CPP), and beam walk (BW) task were evaluated previously and at various times post-trauma. Long-time locomotor and cognitive consequences were measured one day before and on days 3, 7, 14, and 21 after the trauma. RESULTS RU486 eliminated the inhibitory effects of P4 on brain edema and BBB leakage (P<0.05, P<0.001, respectively). RU486 inhibited the decremental effect of P4 on ICP as well as the increasing effect of P4 on CPP (P<0.001) after TBI. Also, RU486 inhibited the effect of P4 on the increase in traversal time and reduction in vestibulomotor score in the BW task (P<0.001). TBI induced motor, cognitive, and anxiety-like disorders, which lasted for 3 weeks after TBI; but, P4 prevented these cognitive and behavioral abnormalities (P<0.05), and RU486 opposed this P4 effect (P<0.001). CONCLUSION The classic progesterone receptors have neuroprotective effects and prevent long-time behavioral and memory deficiency after brain trauma.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Physiology Research Centers, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Navid Ebrahimi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Neda Salmani
- Department of Psychology, Genetic Institute, Islamic Azad University- Zarand Branch, Kerman, Iran
| |
Collapse
|
11
|
Vahidinia Z, Karimian M, Joghataei MT. Neurosteroids and their receptors in ischemic stroke: From molecular mechanisms to therapeutic opportunities. Pharmacol Res 2020; 160:105163. [DOI: 10.1016/j.phrs.2020.105163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
|
12
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
13
|
Khan H, Kashyap A, Kaur A, Singh TG. Pharmacological postconditioning: a molecular aspect in ischemic injury. J Pharm Pharmacol 2020; 72:1513-1527. [PMID: 33460133 DOI: 10.1111/jphp.13336] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/21/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Ischaemia/reperfusion (I/R) injury is defined as the damage to the tissue which is caused when blood supply returns to tissue after ischaemia. To protect the ischaemic tissue from irreversible injury, various protective agents have been studied but the benefits have not been clinically applicable due to monotargeting, low potency, late delivery or poor tolerability. KEY FINDINGS Strategies involving preconditioning or postconditioning can address the issues related to the failure of protective therapies. In principle, postconditioning (PoCo) is clinically more applicable in the conditions in which there is unannounced ischaemic event. Moreover, PoCo is an attractive beneficial strategy as it can be induced rapidly at the onset of reperfusion via series of brief I/R cycles following a major ischaemic event or it can be induced in a delayed manner. Various pharmacological postconditioning (pPoCo) mechanisms have been investigated systematically. Using different animal models, most of the studies on pPoCo have been carried out preclinically. SUMMARY However, there is a need for the optimization of the clinical protocols to quicken pPoCo clinical translation for future studies. This review summarizes the involvement of various receptors and signalling pathways in the protective mechanisms of pPoCo.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Ankita Kashyap
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
14
|
Sara S, Mohammad K, Nader S, Maryam I, Marzieh S, Elham J, Neda S. Using the NGF/IL-6 ratio as a reliable criterion to show the beneficial effects of progesterone after experimental diffuse brain injury. Heliyon 2020; 6:e03844. [PMID: 32373743 PMCID: PMC7191606 DOI: 10.1016/j.heliyon.2020.e03844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/08/2019] [Accepted: 04/21/2020] [Indexed: 01/19/2023] Open
Abstract
Acute progesterone injection has been shown to reduce brain edema following traumatic brain injury (TBI) due to its neuroprotective effect. We investigated the effects of sustained release of progesterone through implantation of subcutaneous capsules on rat's brain edema and alteration of cerebrospinal fluid (CSF), and serum ratio of NGF/IL-6 after TBI. This experiment was performed on ovariectomized (OVX) rats and the brain injury was induced by Marmarou's method. A high and a low dose of progesterone (HP and LP) was injected intraperitoneally two h after the brain injury. In addition, in the capsule progesterone-treated group (CP), the intervention was implemented 6 h after the brain injury. Brain edema, NGF and IL-6 biomarkers in serum and cerebrospinal fluid (CSF) were measured 48 h after the TBI in injection groups and one week after the TBI in the CP group. No significant difference was found in the two groups or in the admonition methods. After TBI, the NGF level increased and IL-6 level decreased by injection of both doses, as well as by taking the capsule. Ratio of NGF/IL-6 in CSF increased significantly by all forms of progesterone administration. The increase in the level of NGF and IL-6 after TBI was higher in CSF than in serum. These results indicated that effects of progesterone in capsule form were better than the injection form. Progesterone probably works by increasing NGF and reducing IL-6. Future studies should investigate the ratio of these biomarkers as a variable to determine the neuroprotective effects of another drug.
Collapse
Affiliation(s)
- Shirazpour Sara
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Khaksari Mohammad
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahrokhi Nader
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Iranpour Maryam
- Pathology and Stem Cell Research Center, Department of Pathology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahryari Marzieh
- Department of Physiology, Neuroscience Research Center, Medical Faculty, Golestan University of Medical Sciences, Gorgan, Iran
| | - Jafari Elham
- Pathology and Stem Cell Research Center, Department of Pathology, Kerman University of Medical Sciences, Kerman, Iran
| | - Salmani Neda
- Department of Psychology, Genetic Institute, Islamic Azad University of Zarand, Keman, Iran
| |
Collapse
|
15
|
Xiao Y, Shen H, Li R, Zhou X, Xiao H, Yan J. A Novel Octapeptide Derived From G Protein-Coupled Receptor 124 Improves Cognitive Function Via Pro-Angiogenesis In A Rat Model Of Chronic Cerebral Hypoperfusion-Induced Vascular Dementia. Drug Des Devel Ther 2019; 13:3669-3682. [PMID: 31695334 PMCID: PMC6815762 DOI: 10.2147/dddt.s226473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/02/2019] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The lack of effective therapies mandates the development of new treatment strategies for vascular dementia (VaD). G protein-coupled receptor 124 (GPR124) may be a therapeutic target for angiogenesis-related diseases of CNS, including VaD. The GCPF peptide is a truncated and screened fragment of the GPR124 extracellular domain. The potential use of GCPF for VaD treatment, angiogenesis and targeting of integrin αvβ3 are evaluated. METHODS AND RESULTS First, the in vivo results indicated that the GCPF peptide could decrease mean escape latency and increase platform crossing times in BCCAO rats. Second, the in vitro and ex vivo results indicated that the GCPF peptide was an active angiogenic peptide and could promote hCMEC/D3 cell migration and adhesion to ECM molecules. Third, in silico analyses predicted that GCPF could specifically interact with integrin αvβ3; the ∆G of GCPF binding to the binding pocket was -16.402 KJ/mol. The molecular characteristics indicated that highly hydrophilic GCPF with a pI of 11.70 had a short half-life in mammals (~1 hr). Finally, the ELISA experiments indicated that low dissociation constant (Kd= 2.412±0.455 nM) corresponds to the high affinity of GCPF for integrin αvβ3. CONCLUSION The data indicate that adhesion of GCPF immobilized on ECM surface to endothelial cells via integrin αvβ3 modulates cellular functions to promote angiogenesis and improve cognitive function. This is the first report to prove that GCPF, a novel octapeptide, may be an effective strategy for VaD therapy.
Collapse
Affiliation(s)
- Ying Xiao
- College of Science, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Hong Shen
- Neuro-Psychiatric Institute, Nanjing Medical University Affiliated Brain Hospital, Nanjing, People’s Republic of China
| | - Rui Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Nanjing, People’s Republic of China
| | - Xia Zhou
- Neuro-Psychiatric Institute, Nanjing Medical University Affiliated Brain Hospital, Nanjing, People’s Republic of China
| | - Hong Xiao
- Neuro-Psychiatric Institute, Nanjing Medical University Affiliated Brain Hospital, Nanjing, People’s Republic of China
| | - Jun Yan
- Department of Geriatric Neurology, Nanjing Medical University Affiliated Brain Hospital, Nanjing, People’s Republic of China
| |
Collapse
|
16
|
Abstract
Cardiovascular ageing and the atherosclerotic process begin very early in life, most likely in utero. They progress over decades of exposure to suboptimal or abnormal metabolic and hormonal risk factors, eventually culminating in very common, costly, and mostly preventable target-organ pathologies, including coronary heart disease, stroke, heart failure, aortic aneurysm, peripheral artery disease, and vascular dementia. In this Review, we discuss findings from preclinical and clinical studies showing that calorie restriction (CR), intermittent fasting, and adjusted diurnal rhythm of feeding, with adequate intake of specific macronutrients and micronutrients, are powerful interventions not only for the prevention of cardiovascular disease but also for slowing the accumulation of molecular damage leading to cardiometabolic dysfunction. Furthermore, we discuss the mechanisms through which a number of other nondietary interventions, such as regular physical activity, mindfulness-based stress-reduction exercises, and some CR-mimetic drugs that target pro-ageing pathways, can potentiate the beneficial effects of a healthy diet in promoting cardiometabolic health.
Collapse
|
17
|
Kobylarek D, Iwanowski P, Lewandowska Z, Limphaibool N, Szafranek S, Labrzycka A, Kozubski W. Advances in the Potential Biomarkers of Epilepsy. Front Neurol 2019; 10:685. [PMID: 31312171 PMCID: PMC6614180 DOI: 10.3389/fneur.2019.00685] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is a group of chronic neurological disorders characterized by recurrent, spontaneous, and unpredictable seizures. It is one of the most common neurological disorders, affecting tens of millions of people worldwide. Comprehensive studies on epilepsy in recent decades have revealed the complexity of epileptogenesis, in which immunological processes, epigenetic modifications, and structural changes in neuronal tissues have been identified as playing a crucial role. This review discusses the recent advances in the biomarkers of epilepsy. We evaluate the possible molecular background underlying the clinical changes observed in recent studies, focusing on therapeutic investigations, and the evidence of their safety and efficacy in the human population. This article reviews the pathophysiology of epilepsy, including recent reports on the effects of oxidative stress and hypoxia, and focuses on specific biomarkers and their clinical implications, along with further perspectives in epilepsy research.
Collapse
Affiliation(s)
- Dominik Kobylarek
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
18
|
Sohrabji F, Okoreeh A, Panta A. Sex hormones and stroke: Beyond estrogens. Horm Behav 2019; 111:87-95. [PMID: 30713101 PMCID: PMC6527470 DOI: 10.1016/j.yhbeh.2018.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
Stroke risk and poor stroke outcomes in postmenopausal women have usually beeen attributed to decreased levels of estrogen. However, two lines of evidence suggest that this hormone may not be solely responsible for elevated stroke risk in this population. First, the increased risk for CVD and stroke occurs much earlier than menopause at a time when estrogen levels are not yet reduced. Second, estrogen therapy has not successfully reduced stroke risk in all studies. Other sex hormones may therefore also contribute to stroke risk. Prior to menopause, levels of the gonadotrophin Follicle Stimulating Hormone (FSH) are elevated while levels of the gonadal peptide inhibin are lowered, indicating an overall decrease in ovarian reserve. Similarly, reduced estrogen levels at menopause significantly increase the ratio of androgens to estrogens. In view of the evidence that androgens may be unfavorable for CVD and stroke, this elevated ratio of testosterone to estrogen may also contribute to the postmenopause-associated stroke risk. This review synthesizes evidence from different clinical populations including natural menopause, surgical menopause, women on chemotherapy, and preclinical stroke models to dissect the role of ovarian hormones and stroke risk and outcomes.
Collapse
Affiliation(s)
- Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, United States of America.
| | - Andre Okoreeh
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, United States of America
| | - Aditya Panta
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX 77807, United States of America
| |
Collapse
|
19
|
Dual Roles of Astrocyte-Derived Factors in Regulation of Blood-Brain Barrier Function after Brain Damage. Int J Mol Sci 2019; 20:ijms20030571. [PMID: 30699952 PMCID: PMC6387062 DOI: 10.3390/ijms20030571] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a major functional barrier in the central nervous system (CNS), and inhibits the extravasation of intravascular contents and transports various essential nutrients between the blood and the brain. After brain damage by traumatic brain injury, cerebral ischemia and several other CNS disorders, the functions of the BBB are disrupted, resulting in severe secondary damage including brain edema and inflammatory injury. Therefore, BBB protection and recovery are considered novel therapeutic strategies for reducing brain damage. Emerging evidence suggests key roles of astrocyte-derived factors in BBB disruption and recovery after brain damage. The astrocyte-derived vascular permeability factors include vascular endothelial growth factors, matrix metalloproteinases, nitric oxide, glutamate and endothelin-1, which enhance BBB permeability leading to BBB disruption. By contrast, the astrocyte-derived protective factors include angiopoietin-1, sonic hedgehog, glial-derived neurotrophic factor, retinoic acid and insulin-like growth factor-1 and apolipoprotein E which attenuate BBB permeability resulting in recovery of BBB function. In this review, the roles of these astrocyte-derived factors in BBB function are summarized, and their significance as therapeutic targets for BBB protection and recovery after brain damage are discussed.
Collapse
|
20
|
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
|
21
|
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: 101] [Impact Index Per Article: 14.4] [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
|
22
|
Shin JA, Jeong SI, Kim HW, Jang G, Ryu DR, Ahn YH, Choi JH, Choi YH, Park EM. Repression of adenosine triphosphate-binding cassette transporter ABCG2 by estrogen increases intracellular glutathione in brain endothelial cells following ischemic reperfusion injury. Neurobiol Aging 2018; 66:138-148. [PMID: 29574357 DOI: 10.1016/j.neurobiolaging.2018.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/31/2018] [Accepted: 02/21/2018] [Indexed: 01/29/2023]
Abstract
The adenosine triphosphate-binding cassette efflux transporter ABCG2, which is located in the blood-brain barrier limits the entry of endogenous compounds and xenobiotics into the brain, and its expression and activity are regulated by estrogen. This study was aimed to define the role of ABCG2 in estrogen-mediated neuroprotection against ischemic injury. ABCG2 protein levels before and after ischemic stroke were increased in the brain of female mice by ovariectomy, which were reversed by estrogen replacement. In brain endothelial cell line bEnd.3, estrogen reduced the basal ABCG2 protein level and efflux activity and protected cells from ischemic injury without inducing ABCG2 expression. When bEnd.3 cells were transfected with ABCG2 small interfering RNA, ischemia-induced cell death was reduced, and the intracellular concentration of glutathione, an antioxidant that is transported by ABCG2, was increased. In addition, after ischemic stroke in ovariectomized mice, estrogen prevented the reduction of intracellular glutathione level in brain microvessels. These data suggested that the suppression of ABCG2 by estrogen is involved in neuroprotection against ischemic injury by increasing intracellular glutathione, and that the modulation of ABCG2 activity offers a therapeutic target for brain diseases in estrogen-deficient aged women.
Collapse
Affiliation(s)
- Jin A Shin
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Sae Im Jeong
- Department of Pharmacology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hye Won Kim
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Gyeonghui Jang
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Dong-Ryeol Ryu
- Department of Internal Medicine, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Young-Ho Ahn
- Department of Molecular Medicine, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ji Ha Choi
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Youn-Hee Choi
- Department of Physiology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 638] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
24
|
Al-Tarrah K, Moiemen N, Lord JM. The influence of sex steroid hormones on the response to trauma and burn injury. BURNS & TRAUMA 2017; 5:29. [PMID: 28920065 PMCID: PMC5597997 DOI: 10.1186/s41038-017-0093-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/19/2017] [Indexed: 12/24/2022]
Abstract
Trauma and related sequelae result in disturbance of homeostatic mechanisms frequently leading to cellular dysfunction and ultimately organ and system failure. Regardless of the type and severity of injury, gender dimorphism in outcomes following trauma have been reported, with females having lower mortality than males, suggesting that sex steroid hormones (SSH) play an important role in the response of body systems to trauma. In addition, several clinical and experimental studies have demonstrated the effects of SSH on the clinical course and outcomes following injury. Animal studies have reported the ability of SSH to modulate immune, inflammatory, metabolic and organ responses following traumatic injury. This indicates that homeostatic mechanisms, via direct and indirect pathways, can be maintained by SSH at local and systemic levels and hence result in more favourable prognosis. Here, we discuss the role and mechanisms by which SSH modulates the response of the body to injury by maintaining various processes and organ functions. Such properties of sex hormones represent potential novel therapeutic strategies and further our understanding of current therapies used following injury such as oxandrolone in burn-injured patients.
Collapse
Affiliation(s)
- K Al-Tarrah
- Institute of Inflammation and Ageing, Birmingham University Medical School, B15 2TT, Birmingham, UK.,Scar Free Foundation Centre for Burns Research, University Hospital Birmingham Foundation Trust, B15 2WB, Birmingham, UK
| | - N Moiemen
- Scar Free Foundation Centre for Burns Research, University Hospital Birmingham Foundation Trust, B15 2WB, Birmingham, UK
| | - J M Lord
- Institute of Inflammation and Ageing, Birmingham University Medical School, B15 2TT, Birmingham, UK
| |
Collapse
|
25
|
Woods SC, May AA, Liu M, Tso P, Begg DP. Using the cerebrospinal fluid to understand ingestive behavior. Physiol Behav 2017; 178:172-178. [PMID: 27923718 PMCID: PMC5944842 DOI: 10.1016/j.physbeh.2016.11.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023]
Abstract
The cerebrospinal fluid (CSF) offers a window into the workings of the brain and blood-brain barrier (BBB). Molecules that enter into the central nervous system (CNS) by passive diffusion or receptor-mediated transport through the choroid plexus often appear in the CSF prior to acting within the brain. Other molecules enter the CNS by passing through the BBB into the brain's interstitial fluid prior to appearing in the CSF. This pattern is also often observed for molecules synthesized by neurons or glia within the CNS. The CSF is therefore an important conduit for the entry and clearance of molecules into/from the CNS and thereby constitutes an important window onto brain activity and barrier function. Assessing the CSF basally, under experimental conditions, or in the context of challenges or metabolic diseases can provide powerful insights about brain function. Here, we review important findings made by our labs, as influenced by the late Randall Sakai, by interrogating the CSF.
Collapse
Affiliation(s)
- Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Aaron A May
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min Liu
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patrick Tso
- Department of Pathology and Molecular Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Denovan P Begg
- School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
26
|
Sadagurski M, Cady G, Miller RA. Anti-aging drugs reduce hypothalamic inflammation in a sex-specific manner. Aging Cell 2017; 16:652-660. [PMID: 28544365 PMCID: PMC5506421 DOI: 10.1111/acel.12590] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2017] [Indexed: 12/22/2022] Open
Abstract
Aging leads to hypothalamic inflammation, but does so more slowly in mice whose lifespan has been extended by mutations that affect GH/IGF‐1 signals. Early‐life exposure to GH by injection, or to nutrient restriction in the first 3 weeks of life, also modulate both lifespan and the pace of hypothalamic inflammation. Three drugs extend lifespan of UM‐HET3 mice in a sex‐specific way: acarbose (ACA), 17‐α‐estradiol (17αE2), and nordihydroguaiaretic acid (NDGA), with more dramatic longevity increases in males in each case. In this study, we examined the effect of these anti‐aging drugs on neuro‐inflammation in hypothalamus and hippocampus. We found that age‐associated hypothalamic inflammation is reduced in males but not in females at 12 months of age by ACA and 17αE2 and at 22 months of age in NDGA‐treated mice. The three drugs blocked indices of hypothalamic reactive gliosis associated with aging, such as Iba‐1‐positive microglia and GFAP‐positive astrocytes, as well as age‐associated overproduction of TNF‐α. This effect was not observed in drug‐treated female mice or in the hippocampus of the drug‐treated animals. On the other hand, caloric restriction (CR; an intervention that extends the lifespan in both sexes) significantly reduced hypothalamic microglia and TNF‐α in both sexes at 12 months of age. Together, these results suggest that the extent of drug‐induced changes in hypothalamic inflammatory processes is sexually dimorphic in a pattern that parallels the effects of these agents on mouse longevity and that mimics the changes seen, in both sexes, of long‐lived nutrient restricted or mutant mice.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Division of Geriatric and Palliative Medicine; Department of Internal Medicine; University of Michigan; Ann Arbor MI USA
| | - Gillian Cady
- Department of Pathology and Geriatrics Center; University of Michigan; Ann Arbor MI USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center; University of Michigan; Ann Arbor MI USA
| |
Collapse
|
27
|
Xiao H, Deng M, Yang B, Tang J, Hu Z. Role of glycogen synthase kinase 3 in ischemia-induced blood-brain barrier disruption in aged female rats. J Neurochem 2017; 142:194-203. [PMID: 28440874 DOI: 10.1111/jnc.14051] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/20/2017] [Accepted: 04/06/2017] [Indexed: 12/19/2022]
Abstract
Estrogen receptors have protective effects against ischemic brain injury. However, the molecular mechanisms underlying this phenomenon have yet to be well studied. Given that inhibition of glycogen synthase kinase (GSK3) can reduce cerebral ischemia/reperfusion injury, we hypothesized that estrogen receptors-mediated protective effects against ischemia-induced blood-brain barrier (BBB) disruption involve inhibition of GSK3. Thus, we evaluated GSK3 expression in the brain of ovariectomized female rats, and examined the effects of intracerebroventricular pre-treatments of SB216763, GSK3 inhibitor, on BBB permeability following middle cerebral artery occlusion (MCAO). We also examined the role of specific estrogen receptor subtype in regulation of GSK3 expression and BBB permeability after MCAO. We found that ovariectomized female rats exhibited increased mRNA levels of estrogen receptor α (ERα) and estrogen receptor β (ERβ), and increased protein levels of GSK3β but not GSK3α in brain cortical areas. Furthermore, intracerebroventricular pre-treatments of SB216763 dose-dependently attenuated brain infarction volume, brain water contents, neurological deficits, and BBB disruption, and increased tight junction protein ZO-1 and occludin expression at 24 h following MCAO. Finally, activation of ERβ but not ERα dose-dependently decreased GSK3β expression at 24 h following MCAO. This was associated with increased tight junction protein expression and improved neurological scores. Thus, our study suggested that activation of ERβ may protect against brain ischemia-induced BBB disruption by inhibiting GSK3β-mediated signaling.
Collapse
Affiliation(s)
- Han Xiao
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mingyang Deng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianguang Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Yao PS, Chen GR, Zheng SF, Kang DZ. Predictors of Postoperative Cerebral Ischemia in Patients with Ruptured Anterior Communicating Artery Aneurysms. World Neurosurg 2017; 103:241-247. [PMID: 28408258 DOI: 10.1016/j.wneu.2017.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Cerebral ischemia is a major contributor to poor outcome after ruptured anterior communicating artery aneurysms (ACoAs), and is not well classified. In this article, we develop a classification and identify risk factors of cerebral ischemia after ruptured ACoAs. METHODS Three hundred sixty patients with ruptured ACoAs undergoing microsurgical clipping were collected. Sex, age, smoking status, Hunt-Hess grade, Fisher grade, hospital stay, surgical timing, hypertension, diabetes, postoperative cerebral ischemia, and postoperative modified Rankin Scale score were collected. Postoperative ischemic changes are classified according to a novel grade (ischemic grade I-IV). RESULTS Predictive factors of postoperative ischemia (grade I-IV) included sex (odds ratio [OR], 1.956; 95% confidence interval [CI], 1.262-3.032; P = 0.003) and Fisher grade (OR, 1.813; 95% CI, 1.144-2.871; P = 0.011). Male sex had a tendency to develop postoperative cerebral ischemia (61.3% in the ischemia group vs. 45.7% in the nonischemia group), while surgical timing did not. However, in patients with postoperative ischemia, early surgery within 3 days (OR, 3.334; 95% CI, 1.411-7.879; P = 0.006) and advanced age greater than 55 years (OR, 2.783; 95% CI, 1.214-6.382; P = 0.016) were risk factors for postoperative neurologic deficits (grade III-IV). CONCLUSIONS Male sex and higher Fisher grade predict postoperative ischemia (grade I-IV), whereas surgical timing does not. However, in patients with postoperative cerebral ischemia, early surgery within 3 days and age greater than 55 years can increase the frequency of postoperative neurological deficits (grade III-IV). Older male patients undergoing early microsurgery had a tendency to develop neurologic deficits.
Collapse
Affiliation(s)
- Pei-Sen Yao
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guo-Rong Chen
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shu-Fa Zheng
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| | - De-Zhi Kang
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
29
|
Wang C, Jiang J, Zhang X, Song L, Sun K, Xu R. Inhibiting HMGB1 Reduces Cerebral Ischemia Reperfusion Injury in Diabetic Mice. Inflammation 2017; 39:1862-1870. [PMID: 27596007 PMCID: PMC5112296 DOI: 10.1007/s10753-016-0418-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
High mobility group box1 (HMGB1) promotes inflammatory injury, and accumulating evidence suggests that it plays a key role in brain ischemia reperfusion (I/R), as well as the development of diabetes mellitus (DM). The purpose of this study was to investigate whether HMGB1 plays a role in brain I/R in a DM mouse model. Diabetes mellitus was induced by a high-calorie diet and streptozotocin treatment, and cerebral ischemia was induced by middle cerebral artery occlusion. We examined HMGB1 levels following cerebral I/R injury in DM and non-DM mice and evaluated the influence of altered HMGB1 levels on the severity of cerebral injury. Serum HMGB1 levels and the inflammatory factors IL-1β, IL-6, and inflammation-related enzyme iNOS were significantly elevated in DM mice with brain I/R compared with non-DM mice with brain I/R. Blocking HMGB1 function by intraperitoneal injection of anti-HMGB1 neutralizing antibodies reversed the inflammatory response and the extent of brain damage, suggesting that HMGB1 plays an important role in cerebral ischemic stroke in diabetic mice.
Collapse
Affiliation(s)
- Chong Wang
- The Military General Hospital of Beijing, PLA, Beijing, 100700 People’s Republic of China
- Jining First People Hospital, Jining, 272011 People’s Republic of China
| | - Jie Jiang
- Jining First People Hospital, Jining, 272011 People’s Republic of China
| | - Xiuping Zhang
- Jinan Central Hospital, Jinan, 250012 People’s Republic of China
| | - Linjie Song
- Jinan Central Hospital, Jinan, 250012 People’s Republic of China
| | - Kai Sun
- Graduate School, Weifang Medical University, Weifang, 261053 People’s Republic of China
| | - Ruxiang Xu
- The Military General Hospital of Beijing, PLA, Beijing, 100700 People’s Republic of China
- Affiliated Bayi Brain Hospital, General Hospital of Beijing, Military Region, No. 5, Nanmencang, Dongcheng District, Beijing, 100000 People’s Republic of China
| |
Collapse
|
30
|
Matrix Metalloproteinase-9 and Recovery of Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2017; 26:733-740. [PMID: 28063771 DOI: 10.1016/j.jstrokecerebrovasdis.2016.09.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 08/17/2016] [Accepted: 09/24/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Stroke outcome can be predicted by clinical features, biochemical parameters, and some risk factors. Matrix metalloproteinase-9 (MMP-9) is involved in various stages of stroke pathology. MMP-9 inhibitors are potential stroke therapeutic agents. Little is known about the relation between MMP-9-after the acute stage-and clinical recovery. OBJECTIVE The study aimed to investigate the serum level of MMP-9 at stroke onset as predictor of stroke outcome and the relation between the level of MMP-9 after 30 days and stroke recovery. METHODS The National Institutes of Health Stroke Scale, modified Rankin Scale, and serum level of MMP-9 were assessed in 30 patients with acute ischemic stroke during the first 24 hours of onset and then a month later. None of the patients received thrombolytic therapy. Thirty normal volunteers of matched age and sex were included in the control group. RESULTS The serum level of MMP-9 at stroke onset was independently positively correlated with stroke outcome. The serum level of MMP-9 30 days after stroke onset was positively correlated with initial stroke severity and outcome, as well as with clinical recovery. CONCLUSION Higher serum level of MMP-9 at stroke onset can be a predictor of poor stroke outcome. However, beyond the acute stage, MMP-9 may play beneficial role in stroke recovery.
Collapse
|
31
|
Strong R, Miller RA, Antebi A, Astle CM, Bogue M, Denzel MS, Fernandez E, Flurkey K, Hamilton KL, Lamming DW, Javors MA, de Magalhães JP, Martinez PA, McCord JM, Miller BF, Müller M, Nelson JF, Ndukum J, Rainger GE, Richardson A, Sabatini DM, Salmon AB, Simpkins JW, Steegenga WT, Nadon NL, Harrison DE. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α-glucosidase inhibitor or a Nrf2-inducer. Aging Cell 2016; 15:872-84. [PMID: 27312235 PMCID: PMC5013015 DOI: 10.1111/acel.12496] [Citation(s) in RCA: 273] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 12/25/2022] Open
Abstract
The National Institute on Aging Interventions Testing Program (ITP) evaluates agents hypothesized to increase healthy lifespan in genetically heterogeneous mice. Each compound is tested in parallel at three sites, and all results are published. We report the effects of lifelong treatment of mice with four agents not previously tested: Protandim, fish oil, ursodeoxycholic acid (UDCA) and metformin - the latter with and without rapamycin, and two drugs previously examined: 17-α-estradiol and nordihydroguaiaretic acid (NDGA), at doses greater and less than used previously. 17-α-estradiol at a threefold higher dose robustly extended both median and maximal lifespan, but still only in males. The male-specific extension of median lifespan by NDGA was replicated at the original dose, and using doses threefold lower and higher. The effects of NDGA were dose dependent and male specific but without an effect on maximal lifespan. Protandim, a mixture of botanical extracts that activate Nrf2, extended median lifespan in males only. Metformin alone, at a dose of 0.1% in the diet, did not significantly extend lifespan. Metformin (0.1%) combined with rapamycin (14 ppm) robustly extended lifespan, suggestive of an added benefit, based on historical comparison with earlier studies of rapamycin given alone. The α-glucosidase inhibitor, acarbose, at a concentration previously tested (1000 ppm), significantly increased median longevity in males and 90th percentile lifespan in both sexes, even when treatment was started at 16 months. Neither fish oil nor UDCA extended lifespan. These results underscore the reproducibility of ITP longevity studies and illustrate the importance of identifying optimal doses in lifespan studies.
Collapse
Affiliation(s)
- Randy Strong
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, Department of PharmacologyThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMI48109‐2200USA
| | - Adam Antebi
- Max Planck Institute for Biology of AgeingCologneD‐50931Germany
| | | | | | | | - Elizabeth Fernandez
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, Department of PharmacologyThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | | | | | - Dudley W. Lamming
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWI53705USA
| | - Martin A. Javors
- Department of PsychiatryUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - João Pedro de Magalhães
- School of Biological SciencesUniversity of LiverpoolCrown StreetLiverpoolL69 7ZBUK
- Present address: Integrative Genomics of Ageing GroupInstitute of Ageing and Chronic DiseaseUniversity of LiverpoolL7 8TX, LiverpoolUnited Kingdom
| | - Paul Anthony Martinez
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, Department of PharmacologyThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Joe M. McCord
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraCOUSA
| | | | - Michael Müller
- Norwich Medical SchoolUniversity of East AngliaNorwichUK
| | - James F. Nelson
- Department of Physiology and Barshop Center for Longevity and Aging StudiesThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | | | - G. Ed. Rainger
- Centre for Cardiovascular SciencesSchool of Clinical and Experimental MedicineThe Medical SchoolThe University of BirminghamBirminghamUK
| | - Arlan Richardson
- Department of Geriatric MedicineUniversity of Oklahoma Health Science CenterOklahoma CityOK73104USA
- VA Medical CenterOklahoma CityOK73104USA
| | - David M. Sabatini
- Whitehead Institute for Biomedical ResearchCambridgeMA02142USA
- Department of BiologyMITCambridgeMA02139USA
- Howard Hughes Medical InstituteMITCambridgeMA02139USA
- Broad Institute of Harvard and MITSeven Cambridge CenterCambridgeMA02142USA
- The David H. Koch Institute for Integrative Cancer Research at MITCambridgeMA02139USA
| | - Adam B. Salmon
- Department of Molecular Medicine and Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - James W. Simpkins
- Center for Basic & Translational Stroke ResearchWest Virginia UniversityMorgantownWV26506USA
| | - Wilma T. Steegenga
- Division of Human NutritionWageningen University and Research CentreWageningenThe Netherlands
| | - Nancy L. Nadon
- Division of Aging BiologyNational Institute on AgingBethesdaMD20892USA
| | | |
Collapse
|
32
|
Haghmorad D, Salehipour Z, Nosratabadi R, Rastin M, Kokhaei P, Mahmoudi MB, Amini AA, Mahmoudi M. Medium-dose estrogen ameliorates experimental autoimmune encephalomyelitis in ovariectomized mice. J Immunotoxicol 2016; 13:885-896. [PMID: 27602995 DOI: 10.1080/1547691x.2016.1223768] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Estrogen is a neuro-protective hormone in various central nervous system (CNS) disorders. The present study evaluated the role of estrogen during experimental autoimmune encephalomyelitis (EAE) at doses selected to mimic any suppressive potential from the hormone during pregnancy. Here, mice were ovariectomized and then 2 weeks later treated with MOG antigen to induce EAE. Concurrently, mice then received (subcutaneously) an implanted pellet to deliver varying estrogen amounts over a 21-day period. Clinical scores and other parameters were monitored daily for the 21 days. At the end of the period, brain/spinal cord histology was performed to measure lymphocyte infiltration; T-cell profiles were determined through ELISA, flow cytometry, and real-time PCR. Transcription factor expression levels in the CNS were assessed using real-time PCR; T-cell differentiation was evaluated via flow cytometry. The results demonstrated that estrogen inhibited development of EAE. Histological studies revealed limited leukocyte infiltration into the CNS. High and medium dose of estrogen increased TH2 and Treg cell production of interleukin (IL)-4, IL-10, and transforming growth factor (TGF)-β, but concurrently resulted in a significant reduction in production of interferon (IFN)-γ, IL-17, and IL-6. Flow cytometry revealed there were also significant decreases in the percentages of TH1 and TH17 cells, as well as significant increase in percentages of Treg and TH2 cells in the spleen and lymph nodes. Real-time PCR results indicated that high- and medium-dose estrogen treatments reduced T-bet and ROR-γt factor expression, but enhanced Foxp3 and GATA3 expression. Collectively, these results demonstrated that a medium dose of estrogen - similar to a pregnancy level of estrogen - could potentially reduce the incidence and severity of autoimmune EAE and possibly other autoimmune pathologies.
Collapse
Affiliation(s)
- Dariush Haghmorad
- a Department of Immunology, School of Medicine , Semnan University of Medical Sciences , Semnan , Iran
| | - Zohreh Salehipour
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Reza Nosratabadi
- c Immunology of Infectious Diseases Research Center , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Maryam Rastin
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Parviz Kokhaei
- a Department of Immunology, School of Medicine , Semnan University of Medical Sciences , Semnan , Iran
| | | | - Abbas Ali Amini
- e Department of Immunology, Faculty of Medicine , Kurdistan University of Medical Sciences , Sanandaj , Iran
| | - Mahmoud Mahmoudi
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
33
|
Lu D, Qu Y, Shi F, Feng D, Tao K, Gao G, He S, Zhao T. Activation of G protein-coupled estrogen receptor 1 (GPER-1) ameliorates blood-brain barrier permeability after global cerebral ischemia in ovariectomized rats. Biochem Biophys Res Commun 2016; 477:209-14. [PMID: 27311857 DOI: 10.1016/j.bbrc.2016.06.044] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/09/2016] [Indexed: 01/21/2023]
Abstract
G protein-coupled estrogen receptor 1 (GPER-1) plays important roles in estrogen-mediated neuroprotection. However, protective effects of GPER-1 on blood-brain barrier (BBB) after ischemic stroke have not been determined. The aim of present study was to determine whether GPER-1 activation ameliorates BBB permeability in ovariectomized rats with induced global cerebral ischemia (GCI). GCI was induced by 4-vessel occlusion for 20 min followed by 24 h reperfusion period. The GPER-1 agonist (G1) was bilaterally administered immediately upon reperfusion by intracerebroventricular (icv) injection. We found that the GPER-1 agonist could significantly decrease immunoglobulin G (IgG) extravasation and increase the levels of tight junctions (occludin and claudin-5) in the CA1 at 24 h of reperfusion after GCI. Further, protein levels of vascular endothelial growth factor A (VEGF-A) was significantly decreased in the ischemic CA1 by G1. Our results suggest that GPER-1 activation reduce tight junctions disruption via inhibition of VEGF-A expression after ischemic injury.
Collapse
Affiliation(s)
- Dan Lu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Fei Shi
- Department of Aerospace Biodynamics, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kai Tao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiming He
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianzhi Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
34
|
Ghanbarabadi M, Iranshahi M, Amoueian S, Mehri S, Motamedshariaty VS, Mohajeri SA. Neuroprotective and memory enhancing effects of auraptene in a rat model of vascular dementia: Experimental study and histopathological evaluation. Neurosci Lett 2016; 623:13-21. [PMID: 27130820 DOI: 10.1016/j.neulet.2016.04.047] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 01/14/2023]
Abstract
Vascular dementia and Alzheimer disease are most common type of dementia. These diseases have been associated with cognitive decline and affected personal behavioral activities. Moreover, the pattern of cerebral blood flow in mild cognitive disorder has appeared as a predictive indication for the development into Alzheimer's disease. Permanent, bilateral occlusion of the common carotid arteries (2VO) is a standard animal model to study vascular dementia and chronic cerebral hypoperfusion. In present study neuroprotective and memory enhancing effects of auraptene (AUR), a citrus coumarin, were studied in 2VO rats. Different doses (25, 8 & 4mg/kg) of AUR were administered orally. The spatial memory performance was tested with Morris water maze after 2VO induction. Biochemical experiments and histopathological evaluations were also applied to investigate the neuroprotective effect of AUR in brain tissue. In comparison with 2VO group, AUR could significantly decrease the scape latency time in treated rats. Also AUR increased the percentage of time spent and traveled pathway in target quadrant on final trial test day. All behavioral results were confirmed by biochemical and histopathological data. Biochemical data indicated that AUR could decrease malondialdehyde (MDA), as lipid peroxidation indicator, and increase glutathione (GSH) content in cortex and hippocampus tissues. Histopathological data showed that AUR could protect cerebrocortical and hippocampus neurons against ischemia. This study demonstrated the memory enhancing effect and neuroprotective activity of AUR after induction of brain ischemia in a rat model of vascular dementia.
Collapse
Affiliation(s)
- Mustafa Ghanbarabadi
- Student Research Committee (SRC), Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sakineh Amoueian
- Pathology department, Imam Reza hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Ahmad Mohajeri
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Abstract
Aging is characterized by the progressive accumulation of degenerative changes, culminating in impaired function and increased probability of death. It is the major risk factor for many human pathologies - including cancer, type 2 diabetes, and cardiovascular and neurodegenerative diseases - and consequently exerts an enormous social and economic toll. The major goal of aging research is to develop interventions that can delay the onset of multiple age-related diseases and prolong healthy lifespan (healthspan). The observation that enhanced longevity and health can be achieved in model organisms by dietary restriction or simple genetic manipulations has prompted the hunt for chemical compounds that can increase lifespan. Most of the pathways that modulate the rate of aging in mammals have homologs in yeast, flies, and worms, suggesting that initial screening to identify such pharmacological interventions may be possible using invertebrate models. In recent years, several compounds have been identified that can extend lifespan in invertebrates, and even in rodents. Here, we summarize the strategies employed, and the progress made, in identifying compounds capable of extending lifespan in organisms ranging from invertebrates to mice and discuss the formidable challenges in translating this work to human therapies.
Collapse
Affiliation(s)
- Surinder Kumar
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David B Lombard
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA; Institute of Gerontology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
36
|
Shin JA, Yoon JC, Kim M, Park EM. Activation of classical estrogen receptor subtypes reduces tight junction disruption of brain endothelial cells under ischemia/reperfusion injury. Free Radic Biol Med 2016; 92:78-89. [PMID: 26784014 DOI: 10.1016/j.freeradbiomed.2016.01.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 01/12/2016] [Accepted: 01/15/2016] [Indexed: 12/15/2022]
Abstract
Ischemic stroke, which induces oxidative stress in the brain, disrupts tight junctions (TJs) between brain endothelial cells, resulting in blood-brain barrier (BBB) breakdown and brain edema. Estrogen reduces oxidative stress and protects brain endothelial cells from ischemic insult. The aim of this study was to determine the protective effects of estrogen on TJ disruption and to examine the roles of classical estrogen receptor (ER) subtypes, ERα- and ERβ, in estrogen effects in brain endothelial cells (bEnd.3) exposed to oxygen-glucose deprivation/reperfusion (OGD/R) injury. Estrogen pretreatment prevented OGD/R-induced decreases in cell viability and TJ protein levels. ERα- and ERβ-specific agonists also reduced TJ disruption. Knockdown of ERα or ERβ expression partially inhibited the effects of estrogen, but completely reversed the effects of corresponding ER subtype-specific agonists on the outcomes of OGD/R. During the early reperfusion period, activation of extracellular signal-regulated kinase1/2 and hypoxia-inducible factor 1α/vascular endothelial growth factor was associated with decreased expression of occludin and claudin-5, respectively, and these changes in TJ protein levels were differentially regulated by ER subtype-specific agonists. Our results suggest that ERα and ERβ activation reduce TJ disruption via inhibition of signaling molecules after ischemic injury and that targeting each ER subtype can be a useful strategy for protecting the BBB from ischemic stroke in postmenopausal women.
Collapse
Affiliation(s)
- Jin A Shin
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 158-710, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Joo Chun Yoon
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Minsuk Kim
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 158-710, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 158-710, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| |
Collapse
|
37
|
Electric stimulation of the ears ameliorated learning and memory impairment in rats with cerebral ischemia-reperfusion injury. Sci Rep 2016; 6:20381. [PMID: 26847826 PMCID: PMC4742903 DOI: 10.1038/srep20381] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/30/2015] [Indexed: 11/28/2022] Open
Abstract
Ear acupuncture enhances the secretion of acetylcholine, which has anti-inflammatory effects. Here we want to investigate the effect of electric stimulation (ES) of the ears on learning and memory impairment in rats with cerebral ischemia-reperfusion injury. At 24 h after reperfusion, 2-Hz ES was applied to the ears for 20 min/day (10 min for each ear) for 7 days continuously. The step-through time of the passive avoidance test was greater in the ES group than in the control group (300.0 ± 0.0 s vs 45.0 ± 26.7 s, p < 0.05). Our results showed that neither neurological deficit score nor motor functions were improved after 2-Hz ES (4.0 ± 0 vs 4.5 ± 0.8, p > 0.05). The numbers of nicotinic acetylcholine receptor α4 positively stained cells in the CA2 and dentate gyrus of the hippocampus were 19.0 ± 11.5 and 269.2 ± 79.3, respectively, in the ES group, which were greater than those in the control group (7.0 ± 5.9 and 165.5 ± 30.8, respectively) (both p < 0.05). These results suggested that 2-Hz ES of the ears ameliorated learning and memory impairment in rats with ischemia-reperfusion injury. ES of the ears has neuroprotective effects, which are related to acetylcholine release.
Collapse
|
38
|
Na W, Lee JY, Kim WS, Yune TY, Ju BG. 17β-Estradiol Ameliorates Tight Junction Disruption via Repression of MMP Transcription. Mol Endocrinol 2015; 29:1347-61. [PMID: 26168035 DOI: 10.1210/me.2015-1124] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) formed by capillary endothelial cells provides a physical wall between the central nervous system (CNS) and circulating blood with highly selective permeability. BBB/BSCB disruption by activation of matrix metalloproteinases (MMPs) has been shown to result in further neurological damage after CNS injury. Recently it has been discovered that estrogen attenuates BBB/BSCB disruption in in vitro and in vivo models. However, the molecular mechanism underlying the estrogen-mediated attenuation of BBB/BSCB disruption has not been elucidated fully. In the present study, we found that 17β-estradiol (E2) suppresses nuclear factor-κB-dependent MMP-1b, MMP-2, MMP-3, MMP-9, MMP-10, and MMP-13 gene activation in microvessel endothelial bEnd.3 cells subjected to oxygen and glucose deprivation/reperfusion injury. E2 induced the recruitment of ERα and nuclear receptor corepressor to the nuclear factor-κB binding site on the MMPs' gene promoters. Consistently, ER antagonist ICI 182.780 showed opposite effects of E2. We further found that E2 attenuates tight junction disruption through the decreased degradation of tight junction proteins in bEnd.3 cells subjected to oxygen and glucose deprivation-reperfusion injury. In addition, E2 suppressed the up-regulation of MMP expression, leading to a decreased BSCB disruption in the injured spinal cord. In conclusion, we discovered the molecular mechanism underlying the protective role of estrogenin BBB/BSCB disruption using an in vitro and in vivo model. Our study suggests that estrogens may provide a potential therapeutic intervention for preserving BBB/BSCB integrity after CNS injury.
Collapse
Affiliation(s)
- Wonho Na
- Department of Life Science (W.N., W.-S.K., B.-G.J.), Sogang University, Seoul 121-742, Korea; and Age-Related and Brain Diseases Research Center (J.Y.L., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Jee Youn Lee
- Department of Life Science (W.N., W.-S.K., B.-G.J.), Sogang University, Seoul 121-742, Korea; and Age-Related and Brain Diseases Research Center (J.Y.L., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Won-Sun Kim
- Department of Life Science (W.N., W.-S.K., B.-G.J.), Sogang University, Seoul 121-742, Korea; and Age-Related and Brain Diseases Research Center (J.Y.L., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Tae Young Yune
- Department of Life Science (W.N., W.-S.K., B.-G.J.), Sogang University, Seoul 121-742, Korea; and Age-Related and Brain Diseases Research Center (J.Y.L., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Bong-Gun Ju
- Department of Life Science (W.N., W.-S.K., B.-G.J.), Sogang University, Seoul 121-742, Korea; and Age-Related and Brain Diseases Research Center (J.Y.L., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
39
|
Lee JY, Choi HY, Na WH, Ju BG, Yune TY. 17β-estradiol inhibits MMP-9 and SUR1/TrpM4 expression and activation and thereby attenuates BSCB disruption/hemorrhage after spinal cord injury in male rats. Endocrinology 2015; 156:1838-50. [PMID: 25763638 DOI: 10.1210/en.2014-1832] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Blood-spinal cord barrier (BSCB) disruption and progressive hemorrhage after spinal cord injury (SCI) lead to secondary injury and the subsequent apoptosis and/or necrosis of neuron and glia, causing permanent neurological deficits. In this study, we examined the effect of 17β-estradiol (E2) on BSCB breakdown and hemorrhage as well as subsequent inflammation after SCI. After a moderate contusion injury at the 9th thoracic segment of spinal cord, E2 (300 μg/kg) was administered by iv injection immediately after SCI, and the same dose of E2 was then administered 6 and 24 hours after injury. Our data show that E2 attenuated BSCB permeability and hemorrhage and reduced the infiltration of neutrophils and macorphages after SCI. Consistent with this finding, the expression of inflammatory mediators was significantly reduced by E2. Furthermore, E2 treatment significantly inhibited the expression of sulfonylurea receptor 1 and transient receptor potential melastatin 4 after injury, which are known to mediate hemorrhage at an early stage after SCI. Moreover, the expression and activation of matrix metalloprotease-9 after injury, which is known to disrupt BSCB, and the degradation of tight junction proteins, such as zona occludens-1 and occludin, were significantly inhibited by E2 treatment. Furthermore, the protective effects of E2 on BSCB disruption and functional improvement were abolished by an estrogen receptor antagonist, ICI 182780 (3 mg/kg). Thus, our study provides evidence that the neuroprotective effect of E2 after SCI is, in part, mediated by inhibiting BSCB disruption and hemorrhage through the down-regulation of sulfonylurea receptor 1/transient receptor potential melastatin 4 and matrix metalloprotease-9, which is dependent on estrogen receptor.
Collapse
Affiliation(s)
- Jee Y Lee
- Age-Related and Brain Diseases Research Center (J.Y.L., H.Y.C., T.Y.Y.) and Department of Biochemistry and Molecular Biology (T.Y.Y.), School of Medicine, Kyung Hee University, Seoul, 130-701, Korea; and Department of Life Science (W.H.N., B.G.J.), Sogang University, Seoul 121-742, Korea
| | | | | | | | | |
Collapse
|
40
|
Sohrabji F. Estrogen-IGF-1 interactions in neuroprotection: ischemic stroke as a case study. Front Neuroendocrinol 2015; 36:1-14. [PMID: 24882635 PMCID: PMC4247812 DOI: 10.1016/j.yfrne.2014.05.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/25/2022]
Abstract
The steroid hormone 17b-estradiol and the peptide hormone insulin-like growth factor (IGF)-1 independently exert neuroprotective actions in neurologic diseases such as stroke. Only a few studies have directly addressed the interaction between the two hormone systems, however, there is a large literature that indicates potentially greater interactions between the 17b-estradiol and IGF-1 systems. The present review focuses on key issues related to this interaction including IGF-1 and sex differences and common activation of second messenger systems. Using ischemic stroke as a case study, this review also focuses on independent and cooperative actions of estrogen and IGF-1 on neuroprotection, blood brain barrier integrity, angiogenesis, inflammation and post-stroke epilepsy. Finally, the review also focuses on the astrocyte, a key mediator of post stroke repair, as a local source of 17b-estradiol and IGF-1. This review thus highlights areas where significant new research is needed to clarify the interactions between these two neuroprotectants.
Collapse
Affiliation(s)
- Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, TAMHSC College of Medicine, Bryan, TX 77807, United States.
| |
Collapse
|
41
|
Zheng Y, Hu Q, Manaenko A, Zhang Y, Peng Y, Xu L, Tang J, Tang J, Zhang JH. 17β-Estradiol attenuates hematoma expansion through estrogen receptor α/silent information regulator 1/nuclear factor-kappa b pathway in hyperglycemic intracerebral hemorrhage mice. Stroke 2014; 46:485-91. [PMID: 25523052 DOI: 10.1161/strokeaha.114.006372] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND PURPOSE 17β-estradiol (E2) has been reported to reduce bleeding and brain injury in experimental intracerebral hemorrhage (ICH) model. However, it is not clear if E2 can prevent early hematoma expansion (HE) induced by hyperglycemia in acute ICH. The aim of this study is to evaluate the effects of E2 on HE and its potential mechanisms in hyperglycemic ICH mice. METHODS Two hundred, 8-week-old male CD1 mice were used. ICH was performed by collagenase injection. 50% dextrose (8 mL/kg) was injected intraperitoneally 3 hours after ICH to induce acute HE (normal saline was used as control). The time course of HE was measured 6, 24, and 72 hours after ICH. Two dosages (100 and 300 μg/kg) of E2 were administrated 1 hour after ICH intraperitoneally. Neurobehavioral deficits, hemorrhage volume, blood glucose level, and blood-brain barrier disruption were measured. To study the mechanisms of E2, estrogen receptor α (ERα) inhibitor methyl-piperidino-pyrazole, silent information regulator 1 (Sirt1) siRNA was administered, respectively. Protein expression of ERα, Sirt1, and acetylated nuclear factor-kappa B, and activity of matrix metalloproteinases-9 were detected. RESULTS Hyperglycemia enhanced HE and deteriorated neurological deficits after ICH from 6 hours after ICH. E2 treatment prevented blood-brain barrier disruption and improved neurological deficits 24 and 72 hours after ICH. E2 reduced HE by activating its receptor ERα, decreasing the expression of Sirt1, deacelylation of nuclear factor-kappa B, and inhibiting the activity of matrix metalloproteinases-9. ERα inhibitor methyl-piperidino-pyrazole and Sirt1 siRNA removed these effects of E2. CONCLUSIONS E2 treatment prevented hyperglycemia-enhanced HE and improved neurological deficits in ICH mice mediated by ERα/Sirt1/nuclear factor-kappa B pathway. E2 may serve as an alternative treatment to decrease early HE after ICH.
Collapse
Affiliation(s)
- Yun Zheng
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Qin Hu
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Anatol Manaenko
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Yang Zhang
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Yan Peng
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Liang Xu
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Junjia Tang
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - Jiping Tang
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng)
| | - John H Zhang
- From the Departments of Physiology and Pharmacology (Y. Zheng, Q.H., A.M., Y. Zhang, Y.P., L.X., Junjia Tang, Jiping Tang, J.H.Z.), and Neurosurgery (J.H.Z.), Loma Linda University School of Medicine, Loma Linda, CA; and Department of Physiology, Medical School of Yangtze University, Jingzhou, Hubei, China (Y. Zheng).
| |
Collapse
|
42
|
Madsen TE, Seigel TA, Mackenzie RS, Marcolini EG, Wira CR, Healy ME, Wright DW, Gentile NT. Gender differences in neurologic emergencies part I: a consensus summary and research agenda on cerebrovascular disease. Acad Emerg Med 2014; 21:1403-13. [PMID: 25422086 DOI: 10.1111/acem.12528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/19/2014] [Accepted: 09/09/2014] [Indexed: 12/12/2022]
Abstract
Cerebrovascular neurologic emergencies including ischemic and hemorrhagic stroke, subarachnoid hemorrhage (SAH), and migraine are leading causes of death and disability that are frequently diagnosed and treated in the emergency department (ED). Although sex and gender differences in neurologic emergencies are beginning to become clearer, there are many unanswered questions about how emergency physicians should incorporate sex and gender into their research initiatives, patient evaluations, and overall management plans for these conditions. After evaluating the existing gaps in the literature, a core group of ED researchers developed a draft of future research priorities. Participants in the 2014 Academic Emergency Medicine consensus conference neurologic emergencies working group then discussed and approved the recommended research agenda using a standardized nominal group technique. Recommendations for future research on the role of sex and gender in the diagnosis, treatment, and outcomes pertinent to ED providers are described for each of three diagnoses: stroke, SAH, and migraine. Recommended future research also includes investigation of the biologic and pathophysiologic differences between men and women with neurologic emergencies as they pertain to ED diagnoses and treatments.
Collapse
Affiliation(s)
- Tracy E. Madsen
- Department of Emergency Medicine; The Alpert Medical School of Brown University Rhode Island Hospital; Providence RI
| | - Todd A. Seigel
- Department of Medicine; UCSF School of Medicine; San Francisco CA
| | | | | | - Charles R. Wira
- Department of Emergency Medicine; Yale University; New Haven CT
| | - Megan E. Healy
- Department of Emergency Medicine; Temple University School of Medicine; Philadelphia PA
| | - David W. Wright
- Department of Emergency Medicine; Emory University School of Medicine; Atlanta GA
| | - Nina T. Gentile
- Department of Emergency Medicine; Temple University School of Medicine; Philadelphia PA
| |
Collapse
|
43
|
Humphreys GI, Ziegler YS, Nardulli AM. 17β-estradiol modulates gene expression in the female mouse cerebral cortex. PLoS One 2014; 9:e111975. [PMID: 25372139 PMCID: PMC4221195 DOI: 10.1371/journal.pone.0111975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/10/2014] [Indexed: 01/13/2023] Open
Abstract
17β-estradiol (E2) plays critical roles in a number of target tissues including the mammary gland, reproductive tract, bone, and brain. Although it is clear that E2 reduces inflammation and ischemia-induced damage in the cerebral cortex, the molecular mechanisms mediating the effects of E2 in this brain region are lacking. Thus, we examined the cortical transcriptome using a mouse model system. Female adult mice were ovariectomized and implanted with silastic tubing containing oil or E2. After 7 days, the cerebral cortices were dissected and RNA was isolated and analyzed using RNA-sequencing. Analysis of the transcriptomes of control and E2-treated animals revealed that E2 treatment significantly altered the transcript levels of 88 genes. These genes were associated with long term synaptic potentiation, myelination, phosphoprotein phosphatase activity, mitogen activated protein kinase, and phosphatidylinositol 3-kinase signaling. E2 also altered the expression of genes linked to lipid synthesis and metabolism, vasoconstriction and vasodilation, cell-cell communication, and histone modification. These results demonstrate the far-reaching and diverse effects of E2 in the cerebral cortex and provide valuable insight to begin to understand cortical processes that may fluctuate in a dynamic hormonal environment.
Collapse
Affiliation(s)
- Gwendolyn I. Humphreys
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yvonne S. Ziegler
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ann M. Nardulli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
44
|
Abstract
Therapeutic hypothermia is the only treatment that has been shown to be of benefit to infant's ≥ 36 weeks of gestation with hypoxic-ischemic encephalopathy. The evidence for the benefit is based on multiple, well-designed randomized clinical trials. Based on this data, the use of therapeutic hypothermia has been widely disseminated throughout the neonatal community. An important concept in hypoxic-ischemic brain injury is the functioning of the neurovascular unit which links neurons, non-neuronal cellular elements and the capillary endothelial cells to promote optimal barrier maintenance between the brain and systemic circulation, regulation of blood flow and neuro-immunologic functioning. Hypoxic-ischemic injury can trigger increased permeability of the blood-brain-barrier via molecular events within the neurovascular unit and initiate pathways to brain injury. In addition, exposure of the brain to cellular elements from the systemic circulation can further propagate the neuro-inflammatory response. The influence of temperature on injury to the neurovascular unit has received relatively little attention. This review will focus on one component of the neurovascular unit, the blood-brain barrier and its constituents. Specifically, this review will address the effects of hypoxia-ischemia and temperature on the neurovascular unit and potential knowledge gaps which may serve as areas for further investigation.
Collapse
Affiliation(s)
- Abbot Laptook
- Warren Alpert Medical School of Brown University, United States; Neonatal Intensive Care Unit, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905, United States.
| |
Collapse
|
45
|
Seker FB, Yorulmaz H, Kaptan E, Caglayan B, Oztas B. Gestational treatment of folic acid attenuates blood-brain barrier leakage in pregnant- and prepubertal rats after pentylenetetrazole-induced seizure. Nutr Neurosci 2014; 19:55-62. [PMID: 25222769 DOI: 10.1179/1476830514y.0000000154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Folic acid (FA) is physiologically important in mammals and is a common vitamin supplement used during pregnancy and lactation. Numerous studies have reported that FA significantly improves endothelial function. The blood-brain barrier (BBB) plays an important role in maintaining the microenvironment required for neuronal function, but its unique structure is damaged by epileptic seizures. The aim of this study was to evaluate the potential protective role of FA on BBB leakage, as well as on the reactive astrogliosis in pregnant rats and their prepubertal offspring during pentylenetetrazole (PTZ)-induced epileptic seizure. METHODS Pregnant rats were treated with FA (5 mg/kg) and PTZ on gestational days 0-19 and 19, respectively. The pups were treated with PTZ at puberty. Evans blue was used to evaluate BBB integrity. Reactive astrogliosis was defined using immunohistochemical analysis for glial fibrillary acidic protein (GFAP). Mean arterial blood pressure (MABP) was measured at the femoral artery. RESULTS A moderate decrease in BBB leakage was observed in FA-treated pregnant and prepubertal animals (P < 0.05). MABP was decreased significantly in pregnant rats (P < 0.05). The epilepsy-induced increase in MABP was less prominent in pregnant animals (P < 0.05). GFAP intensity decreased in PTZ-treated pregnant animals (P < 0.01) and FA-treated prepubertal rats. DISCUSSION Our findings suggest that FA, which is used as a maternal vitamin to promote normal fetus development, may be beneficial against seizure-induced neuronal damage by decreasing BBB leakage and reactive astrogliosis in pregnant and prepubertal rats.
Collapse
|
46
|
Witt KA, Sandoval KE. Steroids and the blood-brain barrier: therapeutic implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:361-390. [PMID: 25307223 DOI: 10.1016/bs.apha.2014.06.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Steroids have a wide spectrum of impact, serving as fundamental regulators of nearly every physiological process within the human body. Therapeutic applications of steroids are equally broad, with a diverse range of medications and targets. Within the central nervous system (CNS), steroids influence development, memory, behavior, and disease outcomes. Moreover, steroids are well recognized as to their impact on the vascular endothelium. The blood-brain barrier (BBB) at the level of the brain microvascular endothelium serves as the principle interface between the peripheral circulation and the brain. Steroids have been identified to impact several critical properties of the BBB, including cellular efflux mechanisms, nutrient uptake, and tight junction integrity. Such actions not only influence brain homeostasis but also the delivery of CNS-targeted therapeutics. A greater understanding of the respective steroid-BBB interactions may shed further light on the differential treatment outcomes observed across CNS pathologies. In this chapter, we examine the current therapeutic implications of steroids respective to BBB structure and function, with emphasis on glucocorticoids and estrogens.
Collapse
Affiliation(s)
- Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University, Edwardsville, Illinois, USA.
| | - Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University, Edwardsville, Illinois, USA
| |
Collapse
|
47
|
Gröger M, Plesnila N. The neuroprotective effect of 17β-estradiol is independent of its antioxidative properties. Brain Res 2014; 1589:61-7. [PMID: 25148707 DOI: 10.1016/j.brainres.2014.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/28/2022]
Abstract
INTRODUCTION 17β-Estradiol (E2) is neuroprotective in experimental models of stroke. While some postulate a mainly antioxidative action due to E2׳s free C3 hydroxyl group at its A-ring, others suggest that neuroprotection is mediated by a hormonal, receptor mediated effect. The aim of the current study was to clarify this issue by testing whether E2 analogues lacking hormonal activity are also neuroprotective following cerebral ischemia. MATERIAL & METHODS Focal cerebral ischemia was induced in male C57/BL6 mice by laser-Doppler-controlled endovascular occlusion of the middle cerebral artery for 40min. Mice received either 1) memantine, a NMDA-receptor antagonist, as a positive control, 2) E2 (1400µg/kg b.w.), or 3) 2,4,6-trimethylphenol (TMP), an E2 analogue without hormonal activity (1400, 140, or 14µg/kg b.w.). Motor function was tested 3h and 24h after ischemia. Thereafter mice were sacrificed and brain damage was quantified by histomorphometry. RESULTS Treatment with memantine or E2 significantly reduced infarct volume by >40% and significantly improved neurological function while treatment with TMP had no effect. CONCLUSION E2 is equally neuroprotective as antagonization of NMDA receptors while E2 analogues without hormonal activity are not neuroprotective. Therefore the current data suggest that the neuroprotection activity of E2 is independent of its free-radical scavenging properties.
Collapse
Affiliation(s)
- Moritz Gröger
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, University of Munich Medical Center, Ludwig-Maximilians-University Munich, Germany; Institute for Surgical Research, University of Munich Medical Center, Ludwig-Maximilians-University Munich, Germany
| | - Nikolaus Plesnila
- Institute for Surgical Research, University of Munich Medical Center, Ludwig-Maximilians-University Munich, Germany; Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig-Maximilians-University Munich, Germany.
| |
Collapse
|
48
|
Harrison DE, Strong R, Allison DB, Ames BN, Astle CM, Atamna H, Fernandez E, Flurkey K, Javors MA, Nadon NL, Nelson JF, Pletcher S, Simpkins JW, Smith D, Wilkinson JE, Miller RA. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 2014; 13:273-82. [PMID: 24245565 PMCID: PMC3954939 DOI: 10.1111/acel.12170] [Citation(s) in RCA: 327] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2013] [Indexed: 01/09/2023] Open
Abstract
Four agents — acarbose (ACA), 17-α-estradiol (EST), nordihydroguaiaretic acid (NDGA), and methylene blue (MB) — were evaluated for lifespan effects in genetically heterogeneous mice tested at three sites. Acarbose increased male median lifespan by 22% (P < 0.0001), but increased female median lifespan by only 5% (P = 0.01). This sexual dimorphism in ACA lifespan effect could not be explained by differences in effects on weight. Maximum lifespan (90th percentile) increased 11% (P < 0.001) in males and 9% (P = 0.001) in females. EST increased male median lifespan by 12% (P = 0.002), but did not lead to a significant effect on maximum lifespan. The benefits of EST were much stronger at one test site than at the other two and were not explained by effects on body weight. EST did not alter female lifespan. NDGA increased male median lifespan by 8–10% at three different doses, with P-values ranging from 0.04 to 0.005. Females did not show a lifespan benefit from NDGA, even at a dose that produced blood levels similar to those in males, which did show a strong lifespan benefit. MB did not alter median lifespan of males or females, but did produce a small, statistically significant (6%, P = 0.004) increase in female maximum lifespan. These results provide new pharmacological models for exploring processes that regulate the timing of aging and late-life diseases, and in particular for testing hypotheses about sexual dimorphism in aging and health.
Collapse
Affiliation(s)
| | - Randy Strong
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Geriatric Research, Education and Clinical Center South Texas Veterans Health Care System San Antonio TX 78229USA
- Research Service South Texas Veterans Health Care System San Antonio TX 78229USA
- Department of Pharmacology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - David B. Allison
- Department of Biostatistics University of Alabama at Birmingham Birmingham AL 35294USA
| | - Bruce N. Ames
- Children's Hospital Oakland Research Institute 5700 Martin Luther King Jr. Way Oakland CA 94609‐1673USA
| | | | - Hani Atamna
- Children's Hospital Oakland Research Institute 5700 Martin Luther King Jr. Way Oakland CA 94609‐1673USA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Geriatric Research, Education and Clinical Center South Texas Veterans Health Care System San Antonio TX 78229USA
- Research Service South Texas Veterans Health Care System San Antonio TX 78229USA
- Department of Pharmacology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | | | - Martin A. Javors
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Department of Psychiatry The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - Nancy L. Nadon
- Division of Aging Biology National Institute on Aging Bethesda MD 20892USA
| | - James F. Nelson
- Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center at San Antonio San Antonio TX 78245USA
- Department of Physiology The University of Texas Health Science Center at San Antonio San Antonio TX 78229USA
| | - Scott Pletcher
- Department of Molecular and Integrative Physiology, and Geriatrics Center University of Michigan Ann Arbor MI 48109USA
| | - James W. Simpkins
- Department of Pharmacology & Neuroscience University of North Texas Health Science Center Fort Worth TX 76107USA
| | - Daniel Smith
- Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL 35294USA
| | - J. Erby Wilkinson
- Unit for Laboratory Animal Medicine University of Michigan School of Medicine Ann Arbor MI 48109USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center University of Michigan Ann Arbor MI 48109USA
| |
Collapse
|
49
|
Cardiotonic pill attenuates white matter and hippocampal damage via inhibiting microglial activation and downregulating ERK and p38 MAPK signaling in chronic cerebral hypoperfused rat. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 13:334. [PMID: 24274593 PMCID: PMC4222777 DOI: 10.1186/1472-6882-13-334] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/22/2013] [Indexed: 11/26/2022]
Abstract
Background The cardiotonic pill (CP) is a herbal medicine composed of Salvia miltiorrhiza (SM), Panax notoginseng (PN), and Dryobalanops aromatica Gaertner (DAG) that is widely used to treat cardiovascular diseases. The present experiment was conducted to examine the effects of CP on white matter and hippocampal damage induced by chronic cerebral hypoperfusion. Methods Chronic cerebral hypoperfusion was induced in male Wistar rats by permanent bilateral common carotid artery occlusion (BCCAo). Daily oral administration of CP (200 mg/kg) began 21 days after BCCAo and continued for 42 days. The levels of microglial activation and myelin basic protein (MBP) were measured in the white matter and hippocampus of rats with chronic BCCAo, and the expression levels of mitogen-activated protein kinases (MAPKs) and inflammatory markers such as cyclooxygenase-2, interleukin-1β, and interleukin-6 were examined. Results MBP expression was reduced in the white matter and hippocampal regions of rats that received BCCAo. In contrast, reduced levels of MBP were not observed in BCCAo rats given CP treatments. The administration of CP alleviated microglial activation, the alteration of ERK and p38 MAPK signaling, and inflammatory mediator expression in rats with chronic BCCAo. Conclusion These results suggest that CP may have protective effects against chronic BCCAo-induced white matter and hippocampal damage by inhibiting inflammatory processes including microglial activation and proinflammatory mediator expression, and downreguating the hyperphosphorylation of ERK and p38 MAPK signaling.
Collapse
|
50
|
Sohrabji F, Bake S, Lewis DK. Age-related changes in brain support cells: Implications for stroke severity. Neurochem Int 2013; 63:291-301. [PMID: 23811611 PMCID: PMC3955169 DOI: 10.1016/j.neuint.2013.06.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/31/2013] [Accepted: 06/19/2013] [Indexed: 12/14/2022]
Abstract
Stroke is one of the leading causes of adult disability and the fourth leading cause of mortality in the US. Stroke disproportionately occurs among the elderly, where the disease is more likely to be fatal or lead to long-term supportive care. Animal models, where the ischemic insult can be controlled more precisely, also confirm that aged animals sustain more severe strokes as compared to young animals. Furthermore, the neuroprotection usually seen in younger females when compared to young males is not observed in older females. The preclinical literature thus provides a valuable resource for understanding why the aging brain is more susceptible to severe infarction. In this review, we discuss the hypothesis that stroke severity in the aging brain may be associated with reduced functional capacity of critical support cells. Specifically, we focus on astrocytes, that are critical for detoxification of the brain microenvironment and endothelial cells, which play a crucial role in maintaining the blood brain barrier. In view of the sex difference in stroke severity, this review also discusses studies of middle-aged acyclic females as well as the effects of the estrogen on astrocytes and endothelial cells.
Collapse
Affiliation(s)
- Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, Texas A&M HSC College of Medicine, Bryan, TX 77807, United States.
| | | | | |
Collapse
|