1
|
Coyoy-Salgado A, Segura-Uribe J, Salgado-Ceballos H, Castillo-Mendieta T, Sánchez-Torres S, Freyermuth-Trujillo X, Orozco-Barrios C, Orozco-Suarez S, Feria-Romero I, Pinto-Almazán R, Moralí de la Brena G, Guerra-Araiza C. Evaluating Sex Steroid Hormone Neuroprotection in Spinal Cord Injury in Animal Models: Is It Promising in the Clinic? Biomedicines 2024; 12:1478. [PMID: 39062051 PMCID: PMC11274729 DOI: 10.3390/biomedicines12071478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The primary mechanism of traumatic spinal cord injury (SCI) comprises the initial mechanical trauma due to the transmission of energy to the spinal cord, subsequent deformity, and persistent compression. The secondary mechanism of injury, which involves structures that remained undamaged after the initial trauma, triggers alterations in microvascular perfusion, the liberation of free radicals and neurotransmitters, lipid peroxidation, alteration in ionic concentrations, and the consequent cell death by necrosis and apoptosis. Research in the treatment of SCI has sought to develop early therapeutic interventions that mitigate the effects of these pathophysiological mechanisms. Clinical and experimental evidence has demonstrated the therapeutic benefits of sex-steroid hormone administration after traumatic brain injury and SCI. The administration of estradiol, progesterone, and testosterone has been associated with neuroprotective effects, better neurological recovery, and decreased mortality after SCI. This review evaluated evidence supporting hormone-related neuroprotection over SCI and the possible underlying mechanisms in animal models. As neuroprotection has been associated with signaling pathways, the effects of these hormones are observed on astrocytes and microglia, modulating the inflammatory response, cerebral blood flow, and metabolism, mediating glutamate excitotoxicity, and their antioxidant effects. Based on the current evidence, it is essential to analyze the benefit of sex steroid hormone therapy in the clinical management of patients with SCI.
Collapse
Affiliation(s)
- Angélica Coyoy-Salgado
- CONAHCyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Julia Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City 06720, Mexico;
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Tzayaka Castillo-Mendieta
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Stephanie Sánchez-Torres
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Ximena Freyermuth-Trujillo
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Carlos Orozco-Barrios
- CONAHCyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Iris Feria-Romero
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City 11340, Mexico
| | - Gabriela Moralí de la Brena
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| |
Collapse
|
2
|
Iqbal J, Huang GD, Xue YX, Yang M, Jia XJ. Role of estrogen in sex differences in memory, emotion and neuropsychiatric disorders. Mol Biol Rep 2024; 51:415. [PMID: 38472517 DOI: 10.1007/s11033-024-09374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Estrogen regulates a wide range of neuronal functions in the brain, such as dendritic spine formation, remodeling of synaptic plasticity, cognition, neurotransmission, and neurodevelopment. Estrogen interacts with intracellular estrogen receptors (ERs) and membrane-bound ERs to produce its effect via genomic and non-genomic pathways. Any alterations in these pathways affect the number, size, and shape of dendritic spines in neurons associated with psychiatric diseases. Increasing evidence suggests that estrogen fluctuation causes changes in dendritic spine density, morphology, and synapse numbers of excitatory and inhibitory neurons differently in males and females. In this review, we discuss the role of estrogen hormone in rodents and humans based on sex differences. First, we explain estrogen role in learning and memory and show that a high estrogen level alleviates the deficits in learning and memory. Secondly, we point out that estrogen produces a striking difference in emotional memories in men and women, which leads them to display sex-specific differences in underlying neuronal signaling. Lastly, we discuss that fluctuations in estrogen levels in men and women are related to neuropsychiatric disorders, including schizophrenia, autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), bipolar disorder (BPD), major depressive disorder (MDD), substance use disorder (SUD), and anxiety disorders.
Collapse
Affiliation(s)
- Javed Iqbal
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Geng-Di Huang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Mei Yang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| | - Xiao-Jian Jia
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
3
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
4
|
Broyles EE, Corell DH, Gidday JM. Maternal repetitive hypoxia prior to mating confers epigenetic resilience to memory impairment in male progeny. Behav Neurosci 2023; 137:178-183. [PMID: 36862475 PMCID: PMC10828958 DOI: 10.1037/bne0000554] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
We showed previously in a mouse model of vascular cognitive impairment and dementia involving chronic cerebral hypoperfusion (CCH) that repetitive hypoxic conditioning (RHC) of both parents results in the epigenetic, intergenerational transmission of resilience to recognition memory loss in adult progeny, as assessed by the novel object recognition test. The present study was undertaken in the same model to determine whether RHC treatment of one or both parents is required to confer dementia resilience intergenerationally. We found inherited resilience to 3 months of CCH in males is maternally mediated (p = .006). Statistically, we observed a strong trend for the paternal germline to contribute as well (p = .052). We also found that, in contrast to what is widely observed in males, females display intact recognition memory (p = .001) after 3 months of CCH, revealing a heretofore unidentified sexual dimorphism with respect to cognitive impact during disease progression. Overall, results of our study strongly implicate epigenetic changes in maternal germ cells, induced by our repetitive systemic hypoxic stimulus, contributing to a modified differentiation program capable of establishing a dementia-resilient phenotype in adult male first-generation progeny. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Emrey E. Broyles
- Department of Ophthalmology, Louisiana State University School of Medicine
| | - David H. Corell
- Department of Ophthalmology, Louisiana State University School of Medicine
| | - Jeffrey M. Gidday
- Department of Ophthalmology, Louisiana State University School of Medicine
- Department of Neuroscience, Louisiana State University School of Medicine
- Department of Physiology, Louisiana State University School of Medicine
- Department of Biochemistry and Molecular Biology, Louisiana State University School of Medicine
| |
Collapse
|
5
|
Abi-Ghanem C, Salinero AE, Kordit D, Mansour FM, Kelly RD, Venkataganesh H, Kyaw NR, Gannon OJ, Riccio D, Fredman G, Poitelon Y, Belin S, Kopec AM, Robison LS, Zuloaga KL. Sex differences in the effects of high fat diet on underlying neuropathology in a mouse model of VCID. Biol Sex Differ 2023; 14:31. [PMID: 37208759 PMCID: PMC10199629 DOI: 10.1186/s13293-023-00513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Damage to the cerebral vasculature can lead to vascular contributions to cognitive impairment and dementia (VCID). A reduction in blood flow to the brain leads to neuropathology, including neuroinflammation and white matter lesions that are a hallmark of VCID. Mid-life metabolic disease (obesity, prediabetes, or diabetes) is a risk factor for VCID which may be sex-dependent (female bias). METHODS We compared the effects of mid-life metabolic disease between males and females in a chronic cerebral hypoperfusion mouse model of VCID. C57BL/6J mice were fed a control or high fat (HF) diet starting at ~ 8.5 months of age. Three months after diet initiation, sham or unilateral carotid artery occlusion surgery (VCID model) was performed. Three months later, mice underwent behavior testing and brains were collected to assess pathology. RESULTS We have previously shown that in this VCID model, HF diet causes greater metabolic impairment and a wider array of cognitive deficits in females compared to males. Here, we report on sex differences in the underlying neuropathology, specifically white matter changes and neuroinflammation in several areas of the brain. White matter was negatively impacted by VCID in males and HF diet in females, with greater metabolic impairment correlating with less myelin markers in females only. High fat diet led to an increase in microglia activation in males but not in females. Further, HF diet led to a decrease in proinflammatory cytokines and pro-resolving mediator mRNA expression in females but not males. CONCLUSIONS The current study adds to our understanding of sex differences in underlying neuropathology of VCID in the presence of a common risk factor (obesity/prediabetes). This information is crucial for the development of effective, sex-specific therapeutic interventions for VCID.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Kordit
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Gabrielle Fredman
- Department Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Yannick Poitelon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Sophie Belin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Ashley M Kopec
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Lisa S Robison
- Department of Psychology & Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL, 33314, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
6
|
Lamorie‐Foote K, Liu Q, Shkirkova K, Ge B, He S, Morgan TE, Mack WJ, Sioutas C, Finch CE, Mack WJ. Particulate matter exposure and chronic cerebral hypoperfusion promote oxidative stress and induce neuronal and oligodendrocyte apoptosis in male mice. J Neurosci Res 2023; 101:384-402. [PMID: 36464774 PMCID: PMC10107949 DOI: 10.1002/jnr.25153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 10/16/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) may amplify the neurotoxicity of nanoscale particulate matter (nPM), resulting in white matter injury. This study characterized the joint effects of nPM (diameter ≤ 200 nm) and CCH secondary to bilateral carotid artery stenosis (BCAS) exposure on neuronal and white matter injury in a murine model. nPM was collected near a highway and re-aerosolized for exposure. Ten-week-old C57BL/6 male mice were randomized into four groups: filtered air (FA), nPM, FA + BCAS, and nPM + BCAS. Mice were exposed to FA or nPM for 10 weeks. BCAS surgeries were performed. Markers of inflammation, oxidative stress, and apoptosis were examined. nPM + BCAS exposure increased brain hemisphere TNFα protein compared to FA. iNOS and HNE immunofluorescence were increased in the corpus callosum and cerebral cortex of nPM + BCAS mice compared to FA. While nPM exposure alone did not decrease cortical neuronal cell count, nPM decreased corpus callosum oligodendrocyte cell count. nPM exposure decreased mature oligodendrocyte cell count and increased oligodendrocyte precursor cell count in the corpus callosum. nPM + BCAS mice exhibited a 200% increase in cortical neuronal TUNEL staining and a 700% increase in corpus callosum oligodendrocyte TUNEL staining compared to FA. There was a supra-additive interaction between nPM and BCAS on cortical neuronal TUNEL staining (2.6× the additive effects of nPM + BCAS). nPM + BCAS exposure increased apoptosis, neuroinflammation, and oxidative stress in the cerebral cortex and corpus callosum. nPM + BCAS exposure increased neuronal apoptosis above the separate responses to each exposure. However, oligodendrocytes in the corpus callosum demonstrated a greater susceptibility to the combined neurotoxic effects of nPM + BCAS exposure.
Collapse
Affiliation(s)
- Krista Lamorie‐Foote
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Neurological Surgery, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Qinghai Liu
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kristina Shkirkova
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Brandon Ge
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Shannon He
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Todd E. Morgan
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Wendy J. Mack
- Department of Population and Public Health SciencesUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Caleb E. Finch
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - William J. Mack
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Neurological Surgery, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
7
|
Belmonte KCD, Holmgren EB, Wills TA, Gidday JM. Epigenetic conditioning induces intergenerational resilience to dementia in a mouse model of vascular cognitive impairment. Alzheimers Dement 2022; 18:1711-1720. [PMID: 35170835 PMCID: PMC9790554 DOI: 10.1002/alz.12616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Epigenetic stimuli induce beneficial or detrimental changes in gene expression, and consequently, phenotype. Some of these phenotypes can manifest across the lifespan-and even in subsequent generations. Here, we used a mouse model of vascular cognitive impairment and dementia (VCID) to determine whether epigenetically induced resilience to specific dementia-related phenotypes is heritable by first-generation progeny. METHODS Our systemic epigenetic therapy consisted of 2 months of repetitive hypoxic "conditioning" (RHC) prior to chronic cerebral hypoperfusion in adult C57BL/6J mice. Resultant changes in object recognition memory and hippocampal long-term potentiation (LTP) were assessed 3 and 4 months later, respectively. RESULTS Hypoperfusion-induced memory/plasticity deficits were abrogated by RHC. Moreover, similarly robust dementia resilience was documented in untreated cerebral hypoperfused animals derived from RHC-treated parents. CONCLUSIONS Our results in experimental VCID underscore the efficacy of epigenetics-based treatments to prevent memory loss, and demonstrate for the first time the heritability of an induced resilience to dementia.
Collapse
Affiliation(s)
- Krystal Courtney D. Belmonte
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Eleanor B. Holmgren
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Tiffany A. Wills
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Jeff M. Gidday
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of Biochemistry and Molecular BiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| |
Collapse
|
8
|
Silva NCBS, Bracko O, Nelson AR, de Oliveira FF, Robison LS, Shaaban CE, Hainsworth AH, Price BR. Vascular cognitive impairment and dementia: An early career researcher perspective. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12310. [PMID: 35496373 PMCID: PMC9043906 DOI: 10.1002/dad2.12310] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023]
Abstract
The field of vascular contributions to cognitive impairment and dementia (VCID) is evolving rapidly. Research in VCID encompasses topics aiming to understand, prevent, and treat the detrimental effects of vascular disease burden in the human brain. In this perspective piece, early career researchers (ECRs) in the field provide an overview of VCID, discuss past and present efforts, and highlight priorities for future research. We emphasize the following critical points as the field progresses: (a) consolidate existing neuroimaging and fluid biomarkers, and establish their utility for pharmacological and non-pharmacological interventions; (b) develop new biomarkers, and new non-clinical models that better recapitulate vascular pathologies; (c) amplify access to emerging biomarker and imaging techniques; (d) validate findings from previous investigations in diverse populations, including those at higher risk of cognitive impairment (e.g., Black, Hispanic, and Indigenous populations); and (e) conduct randomized controlled trials within diverse populations with well-characterized vascular pathologies emphasizing clinically meaningful outcomes.
Collapse
Affiliation(s)
- Nárlon C. Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain HealthDepartment of Physical TherapyFaculty of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Oliver Bracko
- Department of BiologyThe University of MiamiCoral GablesFloridaUSA
| | - Amy R. Nelson
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | | | - Lisa S. Robison
- Department of Psychology and NeuroscienceNova Southeastern UniversityFort LauderdaleFloridaUSA
| | | | - Atticus H. Hainsworth
- Molecular & Clinical Sciences Research InstituteSt George's University of London, UKDepartment of NeurologySt George's University Hospitals NHS Foundation Trust LondonLondonUK
| | - Brittani R. Price
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
9
|
Huuskonen MT, Liu Q, Lamorie-Foote K, Shkirkova K, Connor M, Patel A, Montagne A, Baertsch H, Sioutas C, Morgan TE, Finch CE, Zlokovic BV, Mack WJ. Air Pollution Particulate Matter Amplifies White Matter Vascular Pathology and Demyelination Caused by Hypoperfusion. Front Immunol 2021; 12:785519. [PMID: 34868068 PMCID: PMC8635097 DOI: 10.3389/fimmu.2021.785519] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/01/2021] [Indexed: 12/04/2022] Open
Abstract
Cerebrovascular pathologies are commonly associated with dementia. Because air pollution increases arterial disease in humans and rodent models, we hypothesized that air pollution would also contribute to brain vascular dysfunction. We examined the effects of exposing mice to nanoparticulate matter (nPM; aerodynamic diameter ≤200 nm) from urban traffic and interactions with cerebral hypoperfusion. C57BL/6 mice were exposed to filtered air or nPM with and without bilateral carotid artery stenosis (BCAS) and analyzed by multiparametric MRI and histochemistry. Exposure to nPM alone did not alter regional cerebral blood flow (CBF) or blood brain barrier (BBB) integrity. However, nPM worsened the white matter hypoperfusion (decreased CBF on DSC-MRI) and exacerbated the BBB permeability (extravascular IgG deposits) resulting from BCAS. White matter MRI diffusion metrics were abnormal in mice subjected to cerebral hypoperfusion and worsened by combined nPM+BCAS. Axonal density was reduced equally in the BCAS cohorts regardless of nPM status, whereas nPM exposure caused demyelination in the white matter with or without cerebral hypoperfusion. In summary, air pollution nPM exacerbates cerebrovascular pathology and demyelination in the setting of cerebral hypoperfusion, suggesting that air pollution exposure can augment underlying cerebrovascular contributions to cognitive loss and dementia in susceptible elderly populations.
Collapse
Affiliation(s)
- Mikko T. Huuskonen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Arati Patel
- Department of Neurological Surgery, University of California San Francisco School of Medicine, San Francisco, CA, United States
| | - Axel Montagne
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
10
|
Mun J, Jung J, Park C. Effects of cerebral hypoperfusion on the cerebral white matter: a meta‑analysis. Acta Neurobiol Exp (Wars) 2021; 81:295-306. [PMID: 34672300 DOI: 10.21307/ane-2021-029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Decreased cerebral blood flow (CBF) in aging is known to induce aging‑related cerebral deteriorations, such as neuronal degeneration, white matter (WM) alterations, and vascular deformations. However, the effects of cerebral hypoperfusion on WM alterations remain unclear. This study investigates the relationship between cerebral hypoperfusion and WM total volume changes by assessing the trends in CBF and WM changes by meta‑analysis. In this meta‑analysis, the differences in CBF were compared according to cerebral hypoperfusion type and the effect of cerebral hypoperfusion on the total volume of WM changes in rodents. Using subgroup analysis, 13 studies were evaluated for comparing CBF according to the type of cerebral hypoperfusion; 12 studies were evaluated for comparing the effects of cerebral hypoperfusion on the total volume of WM changes. Our meta‑analysis shows that the total volume of WM decreases with a decrease in CBF. However, the reduction in\r\nthe total volume of WM was greater in normal aging mice than in the cerebral hypoperfusion model mice. These results suggest that the reduction of cerebral WM volume during the aging process is affected by other factors in addition to a decrease in CBF.
Collapse
Affiliation(s)
- Juyeon Mun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Junyang Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chan Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea;
| |
Collapse
|
11
|
Liu Q, Shkirkova K, Lamorie-Foote K, Connor M, Patel A, Babadjouni R, Huuskonen M, Montagne A, Baertsch H, Zhang H, Chen JC, Mack WJ, Walcott BP, Zlokovic BV, Sioutas C, Morgan TE, Finch CE, Mack WJ. Air Pollution Particulate Matter Exposure and Chronic Cerebral Hypoperfusion and Measures of White Matter Injury in a Murine Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:87006. [PMID: 34424052 PMCID: PMC8382048 DOI: 10.1289/ehp8792] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Exposure to ambient air pollution particulate matter (PM) is associated with increased risk of dementia and accelerated cognitive loss. Vascular contributions to cognitive impairment are well recognized. Chronic cerebral hypoperfusion (CCH) promotes neuroinflammation and blood-brain barrier weakening, which may augment neurotoxic effects of PM. OBJECTIVES This study examined interactions of nanoscale particulate matter (nPM; fine particulate matter with aerodynamic diameter ≤ 200 nm ) and CCH secondary to bilateral carotid artery stenosis (BCAS) in a murine model to produce white matter injury. Based on other air pollution interactions, we predicted synergies of nPM with BCAS. METHODS nPM was collected using a particle sampler near a Los Angeles, California, freeway. Mice were exposed to 10 wk of reaerosolized nPM or filtered air (FA) for 150 h. CCH was induced by BCAS surgery. Mice (C57BL/6J males) were randomized to four exposure paradigms: a) FA, b) nPM, c) FA + BCAS , and d) nPM + BCAS . Behavioral outcomes, white matter injury, glial cell activation, inflammation, and oxidative stress were assessed. RESULTS The joint nPM + BCAS group exhibited synergistic effects on white matter injury (2.3× the additive nPM and FA + BCAS scores) with greater loss of corpus callosum volume on T2 magnetic resonance imaging (MRI) (30% smaller than FA group). Histochemical analyses suggested potential microglial-specific inflammatory responses with synergistic effects on corpus callosum C5 immunofluorescent density and whole brain nitrate concentrations (2.1× and 3.9× the additive nPM and FA + BCAS effects, respectively) in the joint exposure group. Transcriptomic responses (RNA-Seq) showed greater impact of nPM + BCAS than individual additive effects, consistent with changes in proinflammatory pathways. Although nPM exposure alone did not alter working memory, the nPM + BCAS cohort demonstrated impaired working memory when compared to the FA + BCAS group. DISCUSSION Our data suggest that nPM and CCH contribute to white matter injury in a synergistic manner in a mouse model. Adverse neurological effects may be aggravated in a susceptible population exposed to air pollution. https://doi.org/10.1289/EHP8792.
Collapse
Affiliation(s)
- Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arati Patel
- Department of Neurological Surgery, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Robin Babadjouni
- Department of Neurological Surgery, Cedars-Sinai, Los Angeles, California, USA
| | - Mikko Huuskonen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Jiu-Chiuan Chen
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Wendy J. Mack
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Brian P. Walcott
- Department of Neurosurgery, Northshore Neurological Institute, Evanston, Illinois, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
12
|
Youssef MI, Ma J, Chen Z, Hu WW. Potential therapeutic agents for ischemic white matter damage. Neurochem Int 2021; 149:105116. [PMID: 34229025 DOI: 10.1016/j.neuint.2021.105116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022]
Abstract
Ischemic white matter damage (WMD) is increasingly being considered as one of the major causes of neurological disorders in older adults and preterm infants. The functional consequences of WMD triggers a progressive cognitive decline and dementia particularly in patients with ischemic cerebrovascular diseases. Despite the major stride made in the pathogenesis mechanisms of ischemic WMD in the last century, effective medications are still not available. So, there is an urgent need to explore a promising approach to slow the progression or modify its pathological course. In this review, we discussed the animal models, the pathological mechanisms and the potential therapeutic agents for ischemic WMD. The development in the studies of anti-oxidants, free radical scavengers, anti-inflammatory or anti-apoptotic agents and neurotrophic factors in ischemic WMD were summarized. The agents which either alleviate oligodendrocyte damage or promote its proliferation or differentiation may have potential value for the treatment of ischemic WMD. Moreover, drugs with multifaceted protective activities or a wide therapeutic window may be optimal for clinical translation.
Collapse
Affiliation(s)
- Mahmoud I Youssef
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Zhong Chen
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Wei-Wei Hu
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
13
|
Xu C, Dai Y, Bai J, Ren B, Xu J, Gao F, Wang L, Zhang W, Wang R. 17β-oestradiol alleviates endoplasmic reticulum stress injury induced by chronic cerebral hypoperfusion through the Haemoglobin/HIF 1α signalling pathway in ovariectomized rats. Neurochem Int 2021; 148:105119. [PMID: 34224805 DOI: 10.1016/j.neuint.2021.105119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/04/2021] [Accepted: 06/27/2021] [Indexed: 11/28/2022]
Abstract
Endoplasmic reticulum stress (ERS) is known to be an essential target in protecting against ischaemic brain injury. In this study, using a vascular dementia (VaD) animal model induced by bilateral common carotid artery occlusion (BCCAO), we evaluated the effect and mechanism of 17β-oestradiol (E2) against VaD by inhibiting ERS at the early stage (14 d, 21 d, 28 d) and late stage (3 m) after BCCAO in the hippocampal CA1 region of ovariectomized rats. The results showed that the activation of the PERK-eIF2α-ATF4-CHOP axis, a typical ERS pathway, was significantly increased at the early and late stages after BCCAO. JNK (c-Jun N-terminal kinase)-cJun, a pro-death pathway, also displayed the same pattern as the ERS axis. E2 treatment profoundly suppressed the impairments caused by BCCAO. Further mechanistic studies revealed that cerebral blood flow (CBF) was sharply decreased at 14 d and returned to the normal level at 21 d after BCCAO. E2 could not change CBF, while it unexpectedly enhanced the ability to carry oxygen. This is evidenced by the fact that the protein expression of haemoglobin α/β (Hα/β), an oxygen carrier, robustly increased at BCCAO 21 d and 3 m after E2 treatment. The oxygen carrier increased strongly after 21 d and 3 m of BCCAO treated with E2. Moreover, E2 correspondingly enhanced the protein expression of hypoxia-inducible factor 1α (HIF 1α) in both the early and late stage after BCCAO in the hippocampal CA1 region. Finally, E2 administration markedly decreased the activities of caspase-8, caspase-3, and caspase-12 and increased the number of NeuN-positive cells. These findings suggest that E2 serves as a neuroprotectant to alleviate VaD by suppressing ERS injury involving the haemoglobin/HIF 1α signalling pathway.
Collapse
Affiliation(s)
- Chao Xu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Yongxin Dai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Bo Ren
- School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Jing Xu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Lu Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Wenli Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; School Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
14
|
Connor M, Lamorie-Foote K, Liu Q, Shkirkova K, Baertsch H, Sioutas C, Morgan TE, Finch CE, Mack WJ. Nanoparticulate matter exposure results in white matter damage and an inflammatory microglial response in an experimental murine model. PLoS One 2021; 16:e0253766. [PMID: 34214084 PMCID: PMC8253444 DOI: 10.1371/journal.pone.0253766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/14/2021] [Indexed: 01/25/2023] Open
Abstract
Exposure to ambient air pollution has been associated with white matter damage and neurocognitive decline. However, the mechanisms of this injury are not well understood and remain largely uncharacterized in experimental models. Prior studies have shown that exposure to particulate matter (PM), a sub-fraction of air pollution, results in neuroinflammation, specifically the upregulation of inflammatory microglia. This study examines white matter and axonal injury, and characterizes microglial reactivity in the corpus callosum of mice exposed to 10 weeks (150 hours) of PM. Nanoscale particulate matter (nPM, aerodynamic diameter ≤200 nm) consisting primarily of traffic-related emissions was collected from an urban area in Los Angeles. Male C57BL/6J mice were exposed to either re-aerosolized nPM or filtered air for 5 hours/day, 3 days/week, for 10 weeks (150 hours; n = 18/group). Microglia were characterized by immunohistochemical double staining of ionized calcium-binding protein-1 (Iba-1) with inducible nitric oxide synthase (iNOS) to identify pro-inflammatory cells, and Iba-1 with arginase-1 (Arg) to identify anti-inflammatory/ homeostatic cells. Myelin injury was assessed by degraded myelin basic protein (dMBP). Oligodendrocyte cell counts were evaluated by oligodendrocyte transcription factor 2 (Olig2). Axonal injury was assessed by axonal neurofilament marker SMI-312. iNOS-expressing microglia were significantly increased in the corpus callosum of mice exposed to nPM when compared to those exposed to filtered air (2.2 fold increase; p<0.05). This was accompanied by an increase in dMBP (1.4 fold increase; p<0.05) immunofluorescent density, a decrease in oligodendrocyte cell counts (1.16 fold decrease; p<0.05), and a decrease in neurofilament SMI-312 (1.13 fold decrease; p<0.05) immunofluorescent density. Exposure to nPM results in increased inflammatory microglia, white matter injury, and axonal degradation in the corpus callosum of adult male mice. iNOS-expressing microglia release cytokines and reactive oxygen/ nitrogen species which may further contribute to the white matter damage observed in this model.
Collapse
Affiliation(s)
- Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States of America
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
15
|
Salinero AE, Robison LS, Gannon OJ, Riccio D, Mansour F, Abi-Ghanem C, Zuloaga KL. Sex-specific effects of high-fat diet on cognitive impairment in a mouse model of VCID. FASEB J 2020; 34:15108-15122. [PMID: 32939871 DOI: 10.1096/fj.202000085r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Mid-life metabolic disease (ie, obesity, diabetes, and prediabetes) causes vascular dysfunction and is a risk factor for vascular contributions to cognitive impairment and dementia (VCID), particularly in women. Using middle-aged mice, we modeled metabolic disease (obesity/prediabetes) via chronic high-fat (HF) diet and modeled VCID via unilateral common carotid artery occlusion. VCID impaired spatial memory in both sexes, but episodic-like memory in females only. HF diet caused greater weight gain and glucose intolerance in middle-aged females than males. HF diet alone impaired episodic-like memory in both sexes, but spatial memory in females only. Finally, the combination of HF diet and VCID elicited cognitive impairments in all tests, in both sexes. Sex-specific correlations were found between metabolic outcomes and memory. Notably, both visceral fat and the pro-inflammatory cytokine tumor necrosis factor alpha correlated with spatial memory deficits in middle-aged females, but not males. Overall, our data show that HF diet causes greater metabolic impairment and a wider array of cognitive deficits in middle-aged females than males. The combination of HF diet with VCID elicits deficits across multiple cognitive domains in both sexes. Our data are in line with clinical data, which shows that mid-life metabolic disease increases VCID risk, particularly in females.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Febronia Mansour
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
16
|
Youssef MI, Zhou Y, Eissa IH, Wang Y, Zhang J, Jiang L, Hu W, Qi J, Chen Z. Tetradecyl 2,3-dihydroxybenzoate alleviates oligodendrocyte damage following chronic cerebral hypoperfusion through IGF-1 receptor. Neurochem Int 2020; 138:104749. [PMID: 32387468 DOI: 10.1016/j.neuint.2020.104749] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Currently, there is no effective therapy for chronic cerebral hypoperfusion-induced subcortical ischemic vascular dementia (SIVD), which displays cognitive deficits and progressive white matter damage. Tetradecyl 2,3-dihydroxybenzoate (ABG-001) is a lead compound derived from gentisides with neuritogenic activity. In this report, we intended to investigate the effect of ABG-001 on the SIVD experimental model through right unilateral common carotid arteries occlusion (rUCCAO) in mice. We found that ABG-001 remarkably alleviated white matter damage and cognitive deficits after cerebral hypoperfusion induced by rUCCAO. The protection of ABG-001 on the white matter was related to an amelioration of the oligodendrocyte apoptosis and demyelination rather than promoting remyelination. Molecular docking study showed that ABG-001 possesses a high affinity for insulin-like growth factor-1 receptor (IGF-1R), but not for tropomyosin receptor kinase A (TrkA). The protection of ABG-001 against oligodendrocyte damage was abrogated by IGF-1R antagonist or knockdown of IGF-1R through shRNA, but not TrkA antagonist. Moreover, ABG-001 did not induce hematological, renal or hepatic toxicity after chronic treatment. The present study indicates that ABG-001 protects oligodendrocytes through IGF-1R to relieve demyelination following chronic cerebral hypoperfusion, which could be represented as an encouraging treatment for SIVD.
Collapse
Affiliation(s)
- Mahmoud I Youssef
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Yiting Zhou
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China; Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, East Qingchun Road 3, Hangzhou, Zhejiang, 310016, PR China
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Yanhui Wang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jing Zhang
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, East Qingchun Road 3, Hangzhou, Zhejiang, 310016, PR China
| | - Lei Jiang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Weiwei Hu
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Jianhua Qi
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
17
|
Toro-Urrego N, Vesga-Jiménez DJ, Herrera MI, Luaces JP, Capani F. Neuroprotective Role of Hypothermia in Hypoxic-ischemic Brain Injury: Combined Therapies using Estrogen. Curr Neuropharmacol 2019; 17:874-890. [PMID: 30520375 PMCID: PMC7052835 DOI: 10.2174/1570159x17666181206101314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/26/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
Hypoxic-ischemic brain injury is a complex network of factors, which is mainly characterized by a decrease in levels of oxygen concentration and blood flow, which lead to an inefficient supply of nutrients to the brain. Hypoxic-ischemic brain injury can be found in perinatal asphyxia and ischemic-stroke, which represent one of the main causes of mortality and morbidity in children and adults worldwide. Therefore, knowledge of underlying mechanisms triggering these insults may help establish neuroprotective treatments. Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators exert several neuroprotective effects, including a decrease of reactive oxygen species, maintenance of cell viability, mitochondrial survival, among others. However, these strategies represent a traditional approach of targeting a single factor of pathology without satisfactory results. Hence, combined therapies, such as the administration of therapeutic hypothermia with a complementary neuroprotective agent, constitute a promising alternative. In this sense, the present review summarizes the underlying mechanisms of hypoxic-ischemic brain injury and compiles several neuroprotective strategies, including Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators, which represent putative agents for combined therapies with therapeutic hypothermia.
Collapse
Affiliation(s)
- Nicolás Toro-Urrego
- Address correspondence to this author at the Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; E-mail:
| | | | | | | | | |
Collapse
|
18
|
Babadjouni R, Patel A, Liu Q, Shkirkova K, Lamorie-Foote K, Connor M, Hodis DM, Cheng H, Sioutas C, Morgan TE, Finch CE, Mack WJ. Nanoparticulate matter exposure results in neuroinflammatory changes in the corpus callosum. PLoS One 2018; 13:e0206934. [PMID: 30395590 PMCID: PMC6218079 DOI: 10.1371/journal.pone.0206934] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/21/2018] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies have established an association between air pollution particulate matter exposure (PM2.5) and neurocognitive decline. Experimental data suggest that microglia play an essential role in air pollution PM-induced neuroinflammation and oxidative stress. This study examined the effect of nano-sized particulate matter (nPM) on complement C5 deposition and microglial activation in the corpus callosum of mice (C57BL/6J males). nPM was collected in an urban Los Angeles region impacted by traffic emissions. Mice were exposed to 10 weeks of re-aerosolized nPM or filtered air for a cumulative 150 hours. nPM-exposed mice exhibited reactive microglia and 2-fold increased local deposition of complement C5/ C5α proteins and complement component C5a receptor 1 (CD88) in the corpus callosum. However, serum C5 levels did not differ between nPM and filtered air cohorts. These findings demonstrate white matter C5 deposition and microglial activation secondary to nPM exposure. The C5 upregulation appears to be localized to the brain.
Collapse
Affiliation(s)
- Robin Babadjouni
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Arati Patel
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Michelle Connor
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Drew M. Hodis
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Hank Cheng
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States of America
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - William J. Mack
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
19
|
Rosenfeld CS, Shay DA, Vieira-Potter VJ. Cognitive Effects of Aromatase and Possible Role in Memory Disorders. Front Endocrinol (Lausanne) 2018; 9:610. [PMID: 30386297 PMCID: PMC6199361 DOI: 10.3389/fendo.2018.00610] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Diverse cognitive functions in many vertebrate species are influenced by local conversion of androgens to 17β-estradiol (E2) by aromatase. This enzyme is highly expressed in various brain regions across species, with some inter-species variation in terms of regional brain expression. Since women with breast cancer and men and women with other disorders are often treated with aromatase inhibitors (AI), these populations might be especially vulnerable to cognitive deficits due to low neuroE2 synthesis, i.e., synthesis of E2 directly within the brain. Animal models have been useful in deciphering aromatase effects on cognitive functions. Consequences of AI administration at various life cycle stages have been assessed on auditory, song processing, and spatial memory in birds and various aspects of cognition in rodent models. Additionally, cognitive deficits have been described in aromatase knockout (ArKO) mice that systemically lack this gene throughout their lifespan. This review will consider evidence to date that AI treatment in male and female rodent models, birds, and humans results in cognitive impairments. How brain aromatase regulates cognitive function throughout the lifespan, and gaps in current knowledge will be considered, along with future directions to better define how aromatase might guide learning and memory from early development through the geriatric period. Better understanding the importance of E2 synthesis on neurobehavioral responses at various ages will likely aid in the discovery of therapeutic strategies to prevent potential cognitive deficits, including Alzheimer's Disease, in individuals treated with AI or those possessing CYP19 gene polymorphisms, as well as cognitive effects of normal aging that may be related to changes in brain aromatase activity.
Collapse
Affiliation(s)
- Cheryl S. Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, United States
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
- *Correspondence: Cheryl S. Rosenfeld
| | - Dusti A. Shay
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Victoria J. Vieira-Potter
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Victoria J. Vieira-Potter
| |
Collapse
|