1
|
Ray JW, Sun X, Cruz‐Diaz N, Pulgar VM, Yamaleyeva LM. Sex differences in middle cerebral artery reactivity and hemodynamics independent from changes in systemic arterial stiffness in Sprague-Dawley rats. Physiol Rep 2025; 13:e70250. [PMID: 40165608 PMCID: PMC11959158 DOI: 10.14814/phy2.70250] [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: 12/30/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
The mechanisms of sex differences in cerebrovascular function are not well understood. In this study, we determined whether sex differences in middle cerebral artery (MCA) reactivity are accompanied with changes in cerebral or systemic arterial resistance and stiffness in young adult Sprague-Dawley (SD) rats. No differences in systolic or diastolic blood pressures were observed between sexes. Heart rate was higher in the female versus male SD. Left MCA pulsatility index (PI) was lower in female versus male SD. No differences in left intracranial internal carotid artery (ICA) PI were observed between sexes. There were no differences in thoracic aorta or left common carotid artery pulse wave velocity (PWV) between sexes. In isolated MCA segments, female left MCA had lower contraction to potassium, but similar maximal contraction and sensitivity to thromboxane A2 receptor agonist U46619. Pre-incubation with indomethacin lowered maximal response and sensitivity to U46619 in male but not female MCA. Endothelial nitric oxide synthase and vascular smooth muscle layer thromboxane A2 receptor immunoreactivity were greater in female versus male SD. We conclude that sex differences in the MCA reactivity are associated with a differential functional profile of MCA in adult SD rats independent from changes in systemic PWV.
Collapse
Affiliation(s)
- Jonathan W. Ray
- Department of Surgery/Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Xuming Sun
- Department of Surgery/Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Nildris Cruz‐Diaz
- Department of Surgery/Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Victor M. Pulgar
- Department of Surgery/Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Pharmaceutical and Clinical SciencesCampbell UniversityBuies CreekNorth CarolinaUSA
| | - Liliya M. Yamaleyeva
- Department of Surgery/Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
2
|
Berends E, Vangrieken P, Amiri N, van de Waarenburg MPH, Scheijen JLJM, Hermes DJHP, Wouters K, van Oostenbrugge RJ, Schalkwijk CG, Foulquier S. Increased Levels of Circulating Methylglyoxal Have No Consequence for Cerebral Microvascular Integrity and Cognitive Function in Young Healthy Mice. Mol Neurobiol 2025; 62:4190-4202. [PMID: 39414727 PMCID: PMC11880179 DOI: 10.1007/s12035-024-04552-3] [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: 03/29/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Diabetes and other age-related diseases are associated with an increased risk of cognitive impairment, but the underlying mechanisms remain poorly understood. Methylglyoxal (MGO), a by-product of glycolysis and a major precursor in the formation of advanced glycation end-products (AGEs), is increased in individuals with diabetes and other age-related diseases and is associated with microvascular dysfunction. We now investigated whether increased levels of circulating MGO can lead to cerebral microvascular dysfunction, blood-brain barrier (BBB) dysfunction, and cognitive impairment. Mice were supplemented or not with 50 mM MGO in drinking water for 13 weeks. Plasma and cortical MGO and MGO-derived AGEs were measured with UPLC-MS/MS. Peripheral and cerebral microvascular integrity and inflammation were investigated. Cerebral blood flow and neurovascular coupling were investigated with laser speckle contrast imaging, and cognitive tests were performed. We found a 2-fold increase in plasma MGO and an increase in MGO-derived AGEs in plasma and cortex. Increased plasma MGO did not lead to cerebral microvascular dysfunction, inflammation, or cognitive decline. This study shows that increased concentrations of plasma MGO are not associated with cerebral microvascular dysfunction and cognitive impairment in healthy mice. Future research should focus on the role of endogenously formed MGO in cognitive impairment.
Collapse
Affiliation(s)
- Eline Berends
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Philippe Vangrieken
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Naima Amiri
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Marjo P H van de Waarenburg
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Jean L J M Scheijen
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Denise J H P Hermes
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
| | - Kristiaan Wouters
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| | - Sébastien Foulquier
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands.
- Faculty of Health Medicine and Life Sciences, Department of Pharmacology and Toxicology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
3
|
Padrela BE, Slivka M, Sneve MH, Garrido PF, Dijsselhof MBJ, Hageman T, Geier O, Grydeland H, Mahroo A, Kuijer JPA, Konstandin S, Eickel K, Barkhof F, Günther M, Walhovd KB, Fjell AM, Mutsaerts HJMM, Petr J. Blood-brain barrier water permeability across the adult lifespan: A multi-echo ASL study. Neurobiol Aging 2025; 147:176-186. [PMID: 39798256 DOI: 10.1016/j.neurobiolaging.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/28/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
An emerging biomarker of blood-brain barrier (BBB) permeability is the time of exchange (Tex) of water from the blood to tissue, as measured by multi-echo arterial spin labeling (ASL) MRI. This new non-invasive sequence, already tested in mice, has recently been adapted to humans and optimized for clinical scanning time. In this study, we studied the normal variability of Tex over age and sex, which needs to be established as a reference for studying changes in neurological disease. We evaluated Tex, cerebral blood flow (CBF) and arterial transit time (ATT) in 209 healthy adults between 26 and 87 years, over age and sex, using general linear models in gray matter, white matter, and regionally in cerebral lobes. After QC, 194 participants were included in the main analysis, and the results demonstrated that both gray matter (GM) and white matter (WM) BBB permeability was higher with higher age (Tex lower by 0.47 ms per year in GM [p < 0.05], and by 0.49 ms in WM, for females; no significant for males), with the largest Tex difference in the frontal lobes (0.64 ms decrease per year, p = 0.011, population average). CBF was lower with higher age in the GM (-0.71 mL/min/100g per year, p < 0.001, for females; -0.31 mL/min/100g per year, p < 0.05, for males). When correcting Tex models for CBF and ATT, effect of age on Tex disappears in the GM, but not in the WM (β=-0.28, p = 0.08). The CBF findings of this study are in line with previous studies, demonstrating the validity of the new sequence. The BBB water permeability variation over age and sex described in this study provides a reference for future BBB research.
Collapse
Affiliation(s)
- Beatriz E Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands.
| | - Maksim Slivka
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Markus H Sneve
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Pablo F Garrido
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mathijs B J Dijsselhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Tamara Hageman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
| | - Oliver Geier
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Håkon Grydeland
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Amnah Mahroo
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Joost P A Kuijer
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Simon Konstandin
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Klaus Eickel
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; mediri GmbH, Heidelberg, Germany; Faculty 1 - Physics / Electrical Engineering, University Bremen, Bremen, Germany
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Henk J M M Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| |
Collapse
|
4
|
Muñoz-Muñoz M, Weston M, Sierra-Ramón M, Bond B, Leal-Martín J, Tomlinson OW, Baltasar-Fernández I, Morín-Martín MM, Losa-Reyna J, Alcazar J, García-García FJ, Ara I. The Influence of Sex, Fitness, BMI, and Cardiovascular Risk Factors on Cerebral Blood Velocity Responsiveness to Graded Exercise Testing in Middle-Aged Adults. J Gerontol A Biol Sci Med Sci 2024; 80:glae257. [PMID: 39835882 DOI: 10.1093/gerona/glae257] [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: 03/01/2024] [Indexed: 01/22/2025] Open
Abstract
Mean middle cerebral artery velocity (MCAv) and the pulsatility index (PI), at rest and in response to exercise, are important markers of cerebrovascular health status in middle-aged adults, when vascular decline assumes substantial relevance. Thus, this study aimed to describe and compare the responses of MCAv and PI to incremental exercise. Two hundred and forty-eight volunteers (50-58 years, 55% women) completed a ramp test on a cycle-ergometer. Gas exchange was assessed on a breath-by-breath basis. MCAv was measured via transcranial Doppler and PI calculated. Cardiovascular disease risk (CVR) factors were determined and comprised of measurements of central obesity, blood pressure, fasted plasma glucose, and lipids. The MCAv and PI responses to exercise were compared across body mass index categories, CVR score, and fitness status. We found sex-specific differences in MCAv and PI at rest. However, both sexes showed a similar relative change to baseline (Δ%MCAvmean). Regarding body mass index, obese women (body mass index > 30 kg m-2) had lower MCAv and Δ%MCAvmean and higher PI compared with normo-weight women during exercise. Apart, women with a 0 CVR score showed higher MCAv and lower PI during exercise than those with a score of +3 CVR. Differences between low and high CVR during exercise in Δ%MCAvmean were also found. Eventually, low fitness showed diminished MCAv and a lower response to exercise than high fitness. This study has highlighted significant variability in MCAv responsiveness to exercise among middle-aged adults. Body composition, CVR, and fitness status may play a significant role in preserving cerebrovascular health. These findings shed light on the importance of understanding the cerebrovascular response to exercise.
Collapse
Affiliation(s)
- Miguel Muñoz-Muñoz
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Max Weston
- Public Health and Sport Sciences, University of Exeter, Exeter, UK
- Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Miguel Sierra-Ramón
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Bert Bond
- Public Health and Sport Sciences, University of Exeter, Exeter, UK
- Exeter Head Impacts, Brain Injury and Trauma (ExHIBIT) Research Group, University of Exeter, Exeter, UK
| | - Javier Leal-Martín
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Owen W Tomlinson
- Public Health and Sport Sciences, University of Exeter, Exeter, UK
| | - Iván Baltasar-Fernández
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- Faculty of Health Sciences, University of Castilla-La Mancha, Talavera de la Reina, Spain
| | - María M Morín-Martín
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Department of Geriatrics, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain
| | - José Losa-Reyna
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Grupo de investigación Valoración del Rendimiento Deportivo, Actividad Física y Salud y Lesiones Deportivas (REDAFLED), Universidad de Valladolid, Soria, Spain
| | - Julian Alcazar
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco José García-García
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Department of Geriatrics, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain
| | - Ignacio Ara
- GENUD Toledo Research Group, Faculty of Sport Sciences, University of Castilla-La Mancha, Toledo, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Edvinsson JCA, Grubor I, Maddahi A, Edvinsson L. Male-female comparison of vasomotor effects of circulating hormones in human intracranial arteries. J Headache Pain 2024; 25:216. [PMID: 39663536 PMCID: PMC11633024 DOI: 10.1186/s10194-024-01933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND The purpose of this study was to examine whether there are sex differences in vasomotor responses and receptor localization of hormones and neuropeptides with relevance to migraine (vasopressin, oxytocin, estrogen, progesterone, testosterone, amylin, adrenomedullin and calcitonin gene-related peptide (CGRP)) in human intracranial arteries. METHODS Human cortical cerebral and middle meningeal arteries were used in this study. The tissues were removed in conjunction with neurosurgery and donated with consent. Vasomotor responses of arteries, after exposure to hormones or neuropeptides, were recorded using a wire myograph. Immunohistochemistry was performed to examine the expression and localization of their receptors within human intracranial arteries. RESULTS Vasopressin showed the strongest contractile responses, followed by oxytocin and progesterone. CGRP displayed the strongest vasodilatory response when compared to adrenomedullin, amylin, testosterone and estrogen. No significant differences were observed in vasomotor responses between male and female arteries. The vasomotor effects were supported by the presence of corresponding receptors in the vascular smooth muscle cells. Estrogen receptors (ERα and ERβ), progesterone receptor (PR), vasopressin 1a receptor (V1aR), and the oxytocin receptor (OTR) were expressed in the walls of both cerebral arteries overlying the cerebral cortex and intracranial arteries of the dura mater. ERα, V1aR, and PR were found to be localized in both smooth muscle cells and endothelium, whereas OTR was exclusively located within the smooth muscle cells. CONCLUSIONS Hypothalamic, sex hormones and the pancreas hormone (amylin) receptors are expressed in the human intracranial artery walls. The vasomotor responses revealed no sex differences, however contractile responses to vasopressin was higher and more potent in MMA compared to CCA when pooling data from both sexes. Overall, the hormones estrogen, progesterone and oxytocin, which drop in circulating levels at onset of menstruation, only showed modest vasomotor responses as compared to CGRP. This suggests that their role in inducing menstrual migraine attacks is not directly related to vasomotor responses.
Collapse
Affiliation(s)
- Jacob C A Edvinsson
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Getingevagen 4, Lund, 22185, Sweden.
| | - Irena Grubor
- Department of Neurosurgery, University Hospital, Lund, Sweden
| | - Aida Maddahi
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Getingevagen 4, Lund, 22185, Sweden
| | - Lars Edvinsson
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Getingevagen 4, Lund, 22185, Sweden
| |
Collapse
|
6
|
Chen B, de Launoit E, Meseguer D, Garcia Caceres C, Eichmann A, Renier N, Schneeberger M. The interactions between energy homeostasis and neurovascular plasticity. Nat Rev Endocrinol 2024; 20:749-759. [PMID: 39054359 DOI: 10.1038/s41574-024-01021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Food intake and energy expenditure are sensed and processed by multiple brain centres to uphold energy homeostasis. Evidence from the past decade points to the brain vasculature as a new critical player in regulating energy balance that functions in close association with the local neuronal networks. Nutritional imbalances alter many properties of the neurovascular system (such as neurovascular coupling and blood-brain barrier permeability), thus suggesting a bidirectional link between the nutritional milieu and neurovascular health. Increasing numbers of people are consuming a Western diet (comprising ultra-processed food with high-fat and high-sugar content) and have a sedentary lifestyle, with these factors contributing to the current obesity epidemic. Emerging pharmacological interventions (for example, glucagon-like peptide 1 receptor agonists) successfully trigger weight loss. However, whether these approaches can reverse the detrimental effects of long-term exposure to the Western diet (such as neurovascular uncoupling, neuroinflammation and blood-brain barrier disruption) and maintain stable body weight in the long-term needs to be clarified in addition to possible adverse effects. Lifestyle interventions revert the nutritional trigger for obesity and positively affect our overall health, including the cardiovascular system. This Perspective examines how lifestyle interventions affect the neurovascular system and neuronal networks.
Collapse
Affiliation(s)
- Bandy Chen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Elisa de Launoit
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - David Meseguer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Cristina Garcia Caceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich & German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, Paris, France
| | - Nicolas Renier
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Marc Schneeberger
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
7
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong E, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-related decline in blood-brain barrier function is more pronounced in males than females in parietal and temporal regions. eLife 2024; 13:RP96155. [PMID: 39495221 PMCID: PMC11534331 DOI: 10.7554/elife.96155] [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] [Indexed: 11/05/2024] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), and shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here, we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8-92 years old, using a non-invasive diffusion-prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60 s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Ophthalmology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brian T Gold
- Department of Neuroscience, College of Medicine, University of KentuckyFrankfortUnited States
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of MedicineBaltimoreUnited States
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell MedicineNew YorkUnited States
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
8
|
Konecny F, Kamar L, Zimmerman I, Whitehead SN, Goldman D, Frisbee JC. Early elevations in arterial pressure: a contributor to rapid depressive symptom emergence in female Zucker rats with metabolic disease? J Appl Physiol (1985) 2024; 137:1324-1340. [PMID: 39359187 PMCID: PMC11573269 DOI: 10.1152/japplphysiol.00586.2024] [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: 07/26/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024] Open
Abstract
One of the growing challenges to public health and clinical outcomes is the emergence of cognitive impairments, particularly depressive symptom severity, because of chronic elevations in metabolic disease and cerebrovascular disease risk. To more clearly delineate these relationships and to assess the potential for sexual dimorphism, we used lean (LZR) and obese Zucker rats (OZR) of increasing age to determine relationships between internal carotid artery (ICA) hemodynamics, cerebral vasculopathies, and the emergence of depressive symptoms. Male OZR exhibited progressive elevations in perfusion pressure within the ICA, which were paralleled by endothelial dysfunction, increased cerebral arterial myogenic activation, and reduced cerebral cortex microvessel density. In contrast, female OZR exhibited a greater degree of ICA hypertension than male OZR but maintained normal endothelial function, myogenic activation, and microvessel density to an older age range than did males. Although both male and female OZR exhibited significant and progressive elevations in depressive symptom severity, these were significantly worse in females. Finally, plasma cortisol concentration was elevated higher and at a younger age in female OZR as compared with males, and this difference was maintained to final animal usage at ∼17 wk of age. These results suggest that an increased severity of blood pressure waves may penetrate the cerebral circulation more deeply in female OZR than in males, which may predispose the females to a more severe emergence of depressive symptoms with chronic metabolic disease, whereas males may be more predisposed to more direct cerebral vasculopathies (e.g., stroke, transient ischemic attack).NEW & NOTEWORTHY We provide novel insight that the superior maintenance of cerebrovascular endothelial function in female versus male rats with chronic metabolic disease buffers myogenic activation of cerebral resistance arteries/arterioles despite worsening hypertension. As hypertension development is earlier and more severe in females, potentially due to an elevated stress response, the blunted myogenic activation allows greater arterial pressure wave penetrance into the cerebral microcirculation and is associated with accelerated emergence/severity of depressive symptoms in obese female rats.
Collapse
Affiliation(s)
- Filip Konecny
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Lujaina Kamar
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Isabel Zimmerman
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Jefferson C Frisbee
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Sabetta Z, Krishna G, Curry-Koski T, Lopez M, Adelson PD, Thomas TC. Sex-dependent temporal changes in astrocyte-vessel interactions following diffuse traumatic brain injury in rats. Front Physiol 2024; 15:1469073. [PMID: 39387100 PMCID: PMC11461938 DOI: 10.3389/fphys.2024.1469073] [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: 07/23/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with diffuse axonal injury (DAI), a primary pathology linked to progressive neurodegeneration and neuroinflammation, including chronic astrogliosis, which influences long-term post-TBI recovery and morbidity. Sex-based differences in blood-brain barrier (BBB) permeability increases the risk of accelerated brain aging and early-onset neurodegeneration. However, few studies have evaluated chronic time course of astrocytic responses around cerebrovascular in the context of aging after TBI and sex dependence. We observed increased glial fibrillary acidic protein (GFAP)-labeled accessory processes branching near and connecting with GFAP-ensheathed cortical vessels, suggesting a critical nuance in astrocyte-vessel interactions after TBI. To quantify this observation, male and female Sprague Dawley rats (∼3 months old, n = 5-6/group) underwent either sham surgery or midline fluid percussion injury. Using immunohistochemical analysis, we quantified GFAP-labeled astrocyte primary and accessory processes that contacted GFAP-ensheathed vessels in the somatosensory barrel cortex at 7, 56, and 168 days post-injury (DPI). TBI significantly increased GFAP-positive primary processes at 7 DPI (P < 0.01) in both sexes. At 56 DPI, these vessel-process interactions remained significantly increased exclusively in males (P < 0.05). At 168 DPI, both sexes showed a significant reduction in vessel-process interactions compared to 7 DPI (P < 0.05); however, a modest but significant injury effect reemerged in females (P < 0.05). A similar sex-dependent pattern in the number of accessory processes provides novel evidence of long-term temporal changes in astrocyte-vessel interactions. TBI-induced changes in astrocyte-vessel interactions may indicate chronic BBB vulnerability and processes responsible for early onset vascular and neurodegenerative pathology.
Collapse
Affiliation(s)
- Zackary Sabetta
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- A.T. Still University Kirksville College of Osteopathic Medicine, Kirksville, MO, United States
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Tala Curry-Koski
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Mackenzie Lopez
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - P. David Adelson
- West Virginia University School of Medicine, Rockefeller Neuroscience Institute, Morgantown, WV, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Phoenix VA Healthcare System, Phoenix, AZ, United States
| |
Collapse
|
10
|
Morais A, Imai T, Jin X, Locascio JJ, Boisserand L, Herman AL, Chauhan A, Lamb J, Nagarkatti K, Diniz MA, Kumskova M, Dhanesha N, Kamat PK, Khan MB, Dhandapani KM, Patel RB, Sutariya B, Shi Y, van Leyen K, Kimberly WT, Hess DC, Aronowski J, Leira EC, Koehler RC, Chauhan AK, Sansing LH, Lyden PD, Ayata C. Biological and Procedural Predictors of Outcome in the Stroke Preclinical Assessment Network (SPAN) Trial. Circ Res 2024; 135:575-592. [PMID: 39034919 PMCID: PMC11428171 DOI: 10.1161/circresaha.123.324139] [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: 12/15/2023] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND The SPAN trial (Stroke Preclinical Assessment Network) is the largest preclinical study testing acute stroke interventions in experimental focal cerebral ischemia using endovascular filament middle cerebral artery occlusion (MCAo). Besides testing interventions against controls, the prospective design captured numerous biological and procedural variables, highlighting the enormous heterogeneity introduced by the multicenter structure that might influence stroke outcomes. Here, we leveraged the unprecedented sample size achieved by the SPAN trial and the prospective design to identify the biological and procedural variables that affect experimental stroke outcomes in transient endovascular filament MCAo. METHODS The study cohort included all mice enrolled and randomized in the SPAN trial (N=1789). Mice were subjected to 60-minute MCAo and followed for a month. Thirteen biological and procedural independent variables and 4 functional (weight loss and 4-point neuroscore on days 1 and 2, corner test on days 7 and 28, and mortality) and 3 tissue (day 2, magnetic resonance imaging infarct volumes and swelling; day 30, magnetic resonance imaging tissue loss) outcome variables were prospectively captured. Multivariable regression with stepwise elimination was used to identify the predictors and their effect sizes. RESULTS Older age, active circadian stage at MCAo, and thinner and longer filament silicone tips predicted higher mortality. Older age, larger body weight, longer anesthesia duration, and longer filament tips predicted worse neuroscores, while high-fat diet and blood flow monitoring predicted milder neuroscores. Older age and a high-fat diet predicted worse corner test performance. While shorter filament tips predicted more ipsiversive turning, longer filament tips appeared to predict contraversive turning. Age, sex, and weight interacted when predicting the infarct volume. Older age was associated with smaller infarcts on day 2 magnetic resonance imaging, especially in animals with larger body weights; this association was most conspicuous in females. High-fat diet also predicted smaller infarcts. In contrast, the use of cerebral blood flow monitoring and more severe cerebral blood flow drop during MCAo, longer anesthesia, and longer filament tips all predicted larger infarcts. Bivariate analyses among the dependent variables highlighted a disconnect between tissue and functional outcomes. CONCLUSIONS Our analyses identified variables affecting endovascular filament MCAo outcome, an experimental stroke model used worldwide. Multiple regression refuted some commonly reported predictors and revealed previously unrecognized associations. Given the multicenter prospective design that represents a sampling of real-world conditions, the degree of heterogeneity mimicking clinical trials, the large number of predictors adjusted for in the multivariable model, and the large sample size, we think this is the most definitive analysis of the predictors of preclinical stroke outcome to date. Future multicenter experimental stroke trials should standardize or at least ensure a balanced representation of the biological and procedural variables identified herein as potential confounders.
Collapse
Affiliation(s)
- Andreia Morais
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Takahiko Imai
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xuyan Jin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Joseph J Locascio
- Harvard Catalyst Biostatistical Consulting Unit, Department of Biostatistics, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| | - Ligia Boisserand
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Alison L. Herman
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Anjali Chauhan
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Karisma Nagarkatti
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Marcio A. Diniz
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mariia Kumskova
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nirav Dhanesha
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Pradip K. Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | | | | | - Rakesh B. Patel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Brijesh Sutariya
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yanrong Shi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Klaus van Leyen
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - W. Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| | - David C. Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Enrique C. Leira
- Departments of Neurology, Neurosurgery, Carver College of Medicine, and Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Raymond C. Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Anil K. Chauhan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Patrick D. Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| |
Collapse
|
11
|
Whitehead BJ, Corbin D, Alexander ML, Bumgarner J, Zhang N, Karelina K, Weil ZM. Cerebral hypoperfusion exacerbates traumatic brain injury in male but not female mice. Eur J Neurosci 2024; 60:4346-4361. [PMID: 38858126 PMCID: PMC11533132 DOI: 10.1111/ejn.16439] [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: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Mild-moderate traumatic brain injuries (TBIs) are prevalent, and while many individuals recover, there is evidence that a significant number experience long-term health impacts, including increased vulnerability to neurodegenerative diseases. These effects are influenced by other risk factors, such as cardiovascular disease. Our study tested the hypothesis that a pre-injury reduction in cerebral blood flow (CBF), mimicking cardiovascular disease, worsens TBI recovery. We induced bilateral carotid artery stenosis (BCAS) and a mild-moderate closed-head TBI in male and female mice, either alone or in combination, and analyzed CBF, spatial learning, memory, axonal damage, and gene expression. Findings showed that BCAS and TBI independently caused a ~10% decrease in CBF. Mice subjected to both BCAS and TBI experienced more significant CBF reductions, notably affecting spatial learning and memory, particularly in males. Additionally, male mice showed increased axonal damage with both BCAS and TBI compared to either condition alone. Females exhibited spatial memory deficits due to BCAS, but these were not worsened by subsequent TBI. Gene expression analysis in male mice highlighted that TBI and BCAS individually altered neuronal and glial profiles. However, the combination of BCAS and TBI resulted in markedly different transcriptional patterns. Our results suggest that mild cerebrovascular impairments, serving as a stand-in for preexisting cardiovascular conditions, can significantly worsen TBI outcomes in males. This highlights the potential for mild comorbidities to modify TBI outcomes and increase the risk of secondary diseases.
Collapse
Affiliation(s)
- Bailey J. Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Megan L. Alexander
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Jacob Bumgarner
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| |
Collapse
|
12
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong LE, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-Related Decline in Blood-Brain Barrier Function is More Pronounced in Males than Females in Parietal and Temporal Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575463. [PMID: 38293052 PMCID: PMC10827081 DOI: 10.1101/2024.01.12.575463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8 to 92 years old, using a non-invasive diffusion prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Ophthalmology, Keck School of Medicine, University of Southern California
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell Medicine
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| |
Collapse
|
13
|
Dinh DD, Wan H, Lidington D, Bolz SS. Female mice display sex-specific differences in cerebrovascular function and subarachnoid haemorrhage-induced injury. EBioMedicine 2024; 102:105058. [PMID: 38490104 PMCID: PMC10955634 DOI: 10.1016/j.ebiom.2024.105058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND In male mice, a circadian rhythm in myogenic reactivity influences the extent of brain injury following subarachnoid haemorrhage (SAH). We hypothesized that female mice have a different cerebrovascular phenotype and consequently, a distinct SAH-induced injury phenotype. METHODS SAH was modelled by pre-chiasmatic blood injection. Olfactory cerebral resistance arteries were functionally assessed by pressure myography; these functional assessments were related to brain histology and neurobehavioral assessments. Cystic fibrosis transmembrane conductance regulator (CFTR) expression was assessed by PCR and Western blot. We compared non-ovariectomized and ovariectomized mice. FINDINGS Cerebrovascular myogenic reactivity is not rhythmic in females and no diurnal differences in SAH-induced injury are observed; ovariectomy does not unmask a rhythmic phenotype for any endpoint. CFTR expression is rhythmic, with similar expression levels compared to male mice. CFTR inhibition studies, however, indicate that CFTR activity is lower in female arteries. Pharmacologically increasing CFTR expression in vivo (3 mg/kg lumacaftor for 2 days) reduces myogenic tone at Zeitgeber time 11, but not Zeitgeber time 23. Myogenic tone is not markedly augmented following SAH in female mice and lumacaftor loses its ability to reduce myogenic tone; nevertheless, lumacaftor confers at least some injury benefit in females with SAH. INTERPRETATION Female mice possess a distinct cerebrovascular phenotype compared to males, putatively due to functional differences in CFTR regulation. This sex difference eliminates the CFTR-dependent cerebrovascular effects of SAH and may alter the therapeutic efficacy of lumacaftor compared to males. FUNDING Brain Aneurysm Foundation, Heart and Stroke Foundation and Ted Rogers Centre for Heart Research.
Collapse
Affiliation(s)
- Danny D Dinh
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, Canada
| | - Hoyee Wan
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, Canada
| | - Darcy Lidington
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, Canada
| | - Steffen-Sebastian Bolz
- Department of Physiology, University of Toronto, Toronto, Canada; Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, Canada; Heart & Stroke / Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Canada.
| |
Collapse
|
14
|
Hart DA. The Heterogeneity of Post-Menopausal Disease Risk: Could the Basis for Why Only Subsets of Females Are Affected Be Due to a Reversible Epigenetic Modification System Associated with Puberty, Menstrual Cycles, Pregnancy and Lactation, and, Ultimately, Menopause? Int J Mol Sci 2024; 25:3866. [PMID: 38612676 PMCID: PMC11011715 DOI: 10.3390/ijms25073866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
For much of human evolution, the average lifespan was <40 years, due in part to disease, infant mortality, predators, food insecurity, and, for females, complications of childbirth. Thus, for much of evolution, many females did not reach the age of menopause (45-50 years of age) and it is mainly in the past several hundred years that the lifespan has been extended to >75 years, primarily due to public health advances, medical interventions, antibiotics, and nutrition. Therefore, the underlying biological mechanisms responsible for disease risk following menopause must have evolved during the complex processes leading to Homo sapiens to serve functions in the pre-menopausal state. Furthermore, as a primary function for the survival of the species is effective reproduction, it is likely that most of the advantages of having such post-menopausal risks relate to reproduction and the ability to address environmental stresses. This opinion/perspective will be discussed in the context of how such post-menopausal risks could enhance reproduction, with improved survival of offspring, and perhaps why such risks are preserved. Not all post-menopausal females exhibit risk for this set of diseases, and those who do develop such diseases do not have all of the conditions. The diseases of the post-menopausal state do not operate as a unified complex, but as independent variables, with the potential for some overlap. The how and why there would be such heterogeneity if the risk factors serve essential functions during the reproductive years is also discussed and the concept of sets of reversible epigenetic changes associated with puberty, pregnancy, and lactation is offered to explain the observations regarding the distribution of post-menopausal conditions and their potential roles in reproduction. While the involvement of an epigenetic system with a dynamic "modification-demodification-remodification" paradigm contributing to disease risk is a hypothesis at this point, validation of it could lead to a better understanding of post-menopausal disease risk in the context of reproduction with commonalities may also lead to future improved interventions to control such risk after menopause.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, and McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
15
|
Lin S, Jiang L, Wei K, Yang J, Cao X, Li C. Sex-Specific Association of Body Mass Index with Hippocampal Subfield Volume and Cognitive Function in Non-Demented Chinese Older Adults. Brain Sci 2024; 14:170. [PMID: 38391744 PMCID: PMC10887390 DOI: 10.3390/brainsci14020170] [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: 01/02/2024] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Recent research suggests a possible association between midlife obesity and an increased risk of dementia in later life. However, the underlying mechanisms remain unclear. Little is known about the relationship between body mass index (BMI) and hippocampal subfield atrophy. In this study, we aimed to explore the associations between BMI and hippocampal subfield volumes and cognitive function in non-demented Chinese older adults. Hippocampal volumes were assessed using structural magnetic resonance imaging. Cognitive function was evaluated using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). A total of 66 participants were included in the final analysis, with 35 females and 31 males. We observed a significant correlation between BMI and the hippocampal fissure volume in older females. In addition, there was a negative association between BMI and the RBANS total scale score, the coding score, and the story recall score, whereas no significant correlations were observed in older males. In conclusion, our findings revealed sex-specific associations between BMI and hippocampal subfield volumes and cognitive performance, providing valuable insights into the development of effective interventions for the early prevention of cognitive decline.
Collapse
Affiliation(s)
- Shaohui Lin
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Department of Geriatrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lijuan Jiang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Kai Wei
- Department of Traditional Chinese Medicine, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 201108, China
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai 201108, China
| | - Junjie Yang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xinyi Cao
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Clinical Neurocognitive Research Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai 200030, China
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai 200030, China
| |
Collapse
|
16
|
Simmons A, Mihalek O, Bimonte Nelson HA, Sirianni RW, Stabenfeldt SE. Acute brain injury and nanomedicine: sex as a biological variable. FRONTIERS IN BIOMATERIALS SCIENCE 2024; 3:1348165. [PMID: 39450372 PMCID: PMC11500709 DOI: 10.3389/fbiom.2024.1348165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Sex as a biological variable has been recognized for decades to be a critical aspect of the drug development process, as differences in drug pharmacology and toxicity in female versus male subjects can drive the success or failure of new therapeutics. These concepts in development of traditional drug systems have only recently begun to be applied for advancing nanomedicine systems that are designed for drug delivery or imaging in the central nervous system (CNS). This review provides a comprehensive overview of the current state of two fields of research - nanomedicine and acute brain injury-centering on sex as a biological variable. We highlight areas of each field that provide foundational understanding of sex as a biological variable in nanomedicine, brain development, immune response, and pathophysiology of traumatic brain injury and stroke. We describe current knowledge on female versus male physiology as well as a growing number of empirical reports that directly address sex as a biological variable in these contexts. In sum, the data make clear two key observations. First, the manner in which sex affects nanomedicine distribution, toxicity, or efficacy is important, complex, and depends on the specific nanoparticle system under considerations; second, although field knowledge is accumulating to enable us to understand sex as a biological variable in the fields of nanomedicine and acute brain injury, there are critical gaps in knowledge that will need to be addressed. We anticipate that understanding sex as a biological variable in the development of nanomedicine systems to treat acute CNS injury will be an important determinant of their success.
Collapse
Affiliation(s)
- Amberlyn Simmons
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Olivia Mihalek
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | | | - Rachael W. Sirianni
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
17
|
Mitchell RHB, Grigorian A, Robertson A, Toma S, Luciw NJ, Karthikeyan S, Mutsaerts HJMM, Fiksenbaum L, Metcalfe AWS, MacIntosh BJ, Goldstein BI. Sex differences in cerebral blood flow among adolescents with bipolar disorder. Bipolar Disord 2024; 26:33-43. [PMID: 37217255 DOI: 10.1111/bdi.13326] [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] [Indexed: 05/24/2023]
Abstract
BACKGROUND Abnormalities in cerebral blood flow (CBF) are common in bipolar disorder (BD). Despite known differences in CBF between healthy adolescent males and females, sex differences in CBF among adolescents with BD have never been studied. OBJECTIVE To examine sex differences in CBF among adolescents with BD versus healthy controls (HC). METHODS CBF images were acquired using arterial spin labeling (ASL) perfusion magnetic resonance imaging (MRI) in 123 adolescents (72 BD: 30M, 42F; 51 HC: 22M, 29F) matched for age (13-20 years). Whole brain voxel-wise analysis was performed in a general linear model with sex and diagnosis as fixed factors, sex-diagnosis interaction effect, and age as a covariate. We tested for main effects of sex, diagnosis, and their interaction. Results were thresholded at cluster forming p = 0.0125, with posthoc Bonferroni correction (p = 0.05/4 groups). RESULTS A main effect of diagnosis (BD > HC) was observed in the superior longitudinal fasciculus (SLF), underlying the left precentral gyrus (F =10.24 (3), p < 0.0001). A main effect of sex (F > M) on CBF was detected in the precuneus/posterior cingulate cortex (PCC), left frontal and occipital poles, left thalamus, left SLF, and right inferior longitudinal fasciculus (ILF). No regions demonstrated a significant sex-by-diagnosis interaction. Exploratory pairwise testing in regions with a main effect of sex revealed greater CBF in females with BD versus HC in the precuneus/PCC (F = 7.1 (3), p < 0.01). CONCLUSION Greater CBF in female adolescents with BD versus HC in the precuneus/PCC may reflect the role of this region in the neurobiological sex differences of adolescent-onset BD. Larger studies targeting underlying mechanisms, such as mitochondrial dysfunction or oxidative stress, are warranted.
Collapse
Affiliation(s)
- Rachel H B Mitchell
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Anahit Grigorian
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Andrew Robertson
- Department of Kinesiology, Research Institute for Aging, University of Waterloo, Ontario, Canada
| | - Simina Toma
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Nicholas J Luciw
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Sudhir Karthikeyan
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Henri J M M Mutsaerts
- Radiology and Nuclear Medicine Vrje Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Lisa Fiksenbaum
- Department of Applied Psychology and Human Development, University of Toronto, Toronto, Ontario, Canada
| | - Arron W S Metcalfe
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program , Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program , Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Benjamin I Goldstein
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Rademeyer KM, R Nass S, Jones AM, Ohene-Nyako M, Hauser KF, McRae M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J Neurovirol 2024; 30:1-21. [PMID: 38280928 PMCID: PMC11232468 DOI: 10.1007/s13365-023-01186-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/29/2024]
Abstract
Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
Collapse
Affiliation(s)
- Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - MaryPeace McRae
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, U.S.A..
| |
Collapse
|
19
|
Bayram E, Coughlin DG, Rajmohan R, Litvan I. Sex differences for clinical correlates of substantia nigra neuron loss in people with Lewy body pathology. Biol Sex Differ 2024; 15:8. [PMID: 38243325 PMCID: PMC10797801 DOI: 10.1186/s13293-024-00583-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Lewy body dementia (LBD) phenotype is associated with the presence and degree of Lewy body, Alzheimer's pathologies, and substantia nigra neuron loss. Nigral neuron loss is associated with parkinsonism in LBD, and females with LBD are less likely than males to have parkinsonism. As sex differences were reported for clinical correlates of Lewy body and Alzheimer's pathologies, we aimed to investigate whether there are also sex differences for correlates of nigral neuron loss. METHODS Data were obtained from the National Alzheimer's Coordinating Center for females (n = 159) and males (n = 263) with brainstem, limbic, and neocortical Lewy body pathology. Sex differences for the nigral neuron loss' association with Lewy body pathology staging and core clinical LBD features (cognitive fluctuations, visual hallucinations, rapid eye movement sleep behavior disorder, parkinsonism) during follow-up were analyzed with generalized linear models adjusting for age and Alzheimer's pathology staging. Whether any of the core clinical features at the time of dementia onset can predict underlying nigral neuron loss for females and males were also analyzed with generalized linear models. RESULTS Compared to males, females died older and had higher levels of Braak tau staging, but had similar levels of Lewy body pathology staging and nigral neuron loss. Females were less likely than males to have a clinical Lewy body disease diagnosis during follow-up. More advanced Lewy body pathology staging was associated with more nigral neuron loss, more so for males than females. More nigral neuron loss was associated with parkinsonism and clinical LBD diagnosis during follow-up, more so for males than females. Across the subgroup with dementia (40 females, 58 males), core LBD features at first visit with dementia were not associated with nigral neuron loss. CONCLUSIONS Nigral neuron loss' association with Lewy body pathology staging and core LBD features can differ by sex. Compared to males, females with Lewy body pathology have a higher risk of underdiagnosis. There is a need to elucidate the mechanisms underlying sex differences for pathology and clinicopathological correlations to advance diagnostic and therapeutic efforts in LBD.
Collapse
Affiliation(s)
- Ece Bayram
- Department of Neurosciences, Parkinson and other Movement Disorders Center, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093-0886, USA.
| | - David G Coughlin
- Department of Neurosciences, Parkinson and other Movement Disorders Center, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093-0886, USA
| | - Ravi Rajmohan
- Department of Neurology, University of California Irvine, 1001 Health Sciences Road, Irvine, CA, 92697-3950, USA
| | - Irene Litvan
- Department of Neurosciences, Parkinson and other Movement Disorders Center, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093-0886, USA
| |
Collapse
|
20
|
Wright ME, Murphy K. A mini-review of the evidence for cerebrovascular changes following gender-affirming hormone replacement therapy and a call for increased focus on cerebrovascular transgender health. Front Hum Neurosci 2023; 17:1303871. [PMID: 38077183 PMCID: PMC10702528 DOI: 10.3389/fnhum.2023.1303871] [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: 09/28/2023] [Accepted: 10/31/2023] [Indexed: 02/12/2024] Open
Abstract
Gender-affirming hormone replacement therapy (gaHRT) is an important step for many in the gender diverse community, associated with increased quality-of-life and lower self-reported scores of depression and anxiety. However, considering the interactions that the involved sex hormones have on vasculature (with oestrogen and testosterone demonstrating vasodilatory and vasoconstricting properties, respectively), it is important for transgender healthcare research to examine how the manipulation of these hormones interact with cerebrovascular structure and functioning. There is a stark lack of research in this area. This mini-review outlines the research suggesting a vascular impact of these sex hormones using evidence from a range of cohorts (e.g., menopause, polycystic ovary syndrome) and discusses the work that has been done into cerebrovascular changes following gaHRT. Finally, recommendations for future research into cerebrovascular health in transgender cohorts following gaHRT are outlined.
Collapse
Affiliation(s)
- Melissa Emily Wright
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
21
|
Cifù A, Janes F, Mio C, Domenis R, Pessa ME, Garbo R, Curcio F, Valente M, Fabris M. Brain Endothelial Cells Activate Neuroinflammatory Pathways in Response to Early Cerebral Small Vessel Disease (CSVD) Patients' Plasma. Biomedicines 2023; 11:3055. [PMID: 38002055 PMCID: PMC10669613 DOI: 10.3390/biomedicines11113055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The pathogenesis of cerebral small vessel disease (CSVD) is largely unknown. Endothelial disfunction has been suggested as the turning point in CSVD development. In this study, we tested the effect of plasma from CSVD patients on human cerebral microvascular endothelial cells with the aim of describing the pattern of endothelial activation. Plasma samples from three groups of young subjects have been tested: PTs (subjects affected by early stage CSVD); CTRLs (control subjects without abnormalities at MRI scanning); BDs (blood donors). Human Brain Endothelial Cells 5i (HBEC5i) were treated with plasma and total RNA was extracted. RNAs were pooled to reduce gene expression-based variability and NGS analysis was performed. Differentially expressed genes were highlighted comparing PTs, CTRLs and BDs with HBEC5i untreated cells. No significantly altered pathway was evaluated in BD-related treatment. Regulation of p38 MAPK cascade (GO:1900744) was the only pathway altered in CTRL-related treatment. Indeed, 36 different biological processes turned out to be deregulated after PT treatment of HBEC5i, i.e., the cytokine-mediated signaling pathway (GO:0019221). Endothelial cells activate inflammatory pathways in response to stimuli from CSVD patients' plasma, suggesting the pathogenetic role of neuroinflammation from the early asymptomatic phases of cerebrovascular disease.
Collapse
Affiliation(s)
- Adriana Cifù
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Francesco Janes
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Catia Mio
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Rossana Domenis
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
| | - Maria Elena Pessa
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Riccardo Garbo
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
- Neurology Unit of Gorizia-Monfalcone, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), 34100 Gorizia, Italy
| | - Francesco Curcio
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| | - Mariarosaria Valente
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Department of Head, Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (M.E.P.); (R.G.)
| | - Martina Fabris
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.C.); (C.M.); (R.D.); (F.C.); (M.V.); (M.F.)
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| |
Collapse
|
22
|
Nagy D, Hricisák L, Walford GP, Lékai Á, Karácsony G, Várbíró S, Ungvári Z, Benyó Z, Pál É. Disruption of Vitamin D Signaling Impairs Adaptation of Cerebrocortical Microcirculation to Carotid Artery Occlusion in Hyperandrogenic Female Mice. Nutrients 2023; 15:3869. [PMID: 37764653 PMCID: PMC10534509 DOI: 10.3390/nu15183869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Vitamin D deficiency contributes to the pathogenesis of age-related cerebrovascular diseases, including ischemic stroke. Sex hormonal status may also influence the prevalence of these disorders, indicated by a heightened vulnerability among postmenopausal and hyperandrogenic women. To investigate the potential interaction between sex steroids and disrupted vitamin D signaling in the cerebral microcirculation, we examined the cerebrovascular adaptation to unilateral carotid artery occlusion (CAO) in intact, ovariectomized, and hyperandrogenic female mice with normal or functionally inactive vitamin D receptor (VDR). We also analyzed the morphology of leptomeningeal anastomoses, which play a significant role in the compensation. Ablation of VDR by itself did not impact the cerebrocortical adaptation to CAO despite the reduced number of pial collaterals. While ovariectomy did not undermine compensatory mechanisms following CAO, androgen excess combined with VDR inactivity resulted in prolonged hypoperfusion in the cerebral cortex ipsilateral to the occlusion. These findings suggest that the cerebrovascular consequences of disrupted VDR signaling are less pronounced in females, providing a level of protection even after ovariectomy. Conversely, even short-term androgen excess with lacking VDR signaling may lead to unfavorable outcomes of ischemic stroke, highlighting the complex interplay between sex steroids and vitamin D in terms of cerebrovascular diseases.
Collapse
Affiliation(s)
- Dorina Nagy
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Guillaume Peter Walford
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Ágnes Lékai
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Gábor Karácsony
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary;
- Department of Obstetrics and Gynecology, University of Szeged, 6725 Szeged, Hungary
- Workgroup for Science Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
| | - Zoltán Ungvári
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Éva Pál
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
23
|
Badji A, Youwakim J, Cooper A, Westman E, Marseglia A. Vascular cognitive impairment - Past, present, and future challenges. Ageing Res Rev 2023; 90:102042. [PMID: 37634888 DOI: 10.1016/j.arr.2023.102042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Vascular cognitive impairment (VCI) is a lifelong process encompassing a broad spectrum of cognitive disorders, ranging from subtle or mild deficits to prodromal and fully developed dementia, originating from cerebrovascular lesions such as large and small vessel disease. Genetic predisposition and environmental exposure to risk factors such as unhealthy lifestyles, hypertension, cardiovascular disease, and metabolic disorders will synergistically interact, yielding biochemical and structural brain changes, ultimately culminating in VCI. However, little is known about the pathological processes underlying VCI and the temporal dynamics between risk factors and disease mechanisms (biochemical and structural brain changes). This narrative review aims to provide an evidence-based summary of the link between individual vascular risk/disorders and cognitive dysfunction and the potential structural and biochemical pathophysiological processes. We also discuss some key challenges for future research on VCI. There is a need to shift from individual risk factors/disorders to comorbid vascular burden, identifying and integrating imaging and fluid biomarkers, implementing a life-course approach, considering possible neuroprotective influences of positive life exposures, and addressing biological sex at birth and gender differences. Finally, this review highlights the need for future researchers to leverage and integrate multidimensional data to advance our understanding of the mechanisms and pathophysiology of VCI.
Collapse
Affiliation(s)
- Atef Badji
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Youwakim
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage (CIRCA), Montreal, QC, Canada; Groupe de Recherche sur la Signalisation Neuronal et la Circuiterie (SNC), Montreal, QC, Canada
| | - Alexandra Cooper
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Anna Marseglia
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Kovalska M, Hnilicova P, Kalenska D, Adamkov M, Kovalska L, Lehotsky J. Alzheimer's Disease-like Pathological Features in the Dorsal Hippocampus of Wild-Type Rats Subjected to Methionine-Diet-Evoked Mild Hyperhomocysteinaemia. Cells 2023; 12:2087. [PMID: 37626897 PMCID: PMC10453870 DOI: 10.3390/cells12162087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/27/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Multifactorial interactions, including nutritional state, likely participate in neurodegeneration's pathogenesis and evolution. Dysregulation in methionine (Met) metabolism could lead to the development of hyperhomocysteinaemia (hHcy), playing an important role in neuronal dysfunction, which could potentially lead to the development of Alzheimer's disease (AD)-like pathological features. This study combines proton magnetic resonance spectroscopy (1H MRS) with immunohistochemical analysis to examine changes in the metabolic ratio and histomorphological alterations in the dorsal rat hippocampus (dentate gyrus-DG) subjected to a high Met diet. Male Wistar rats (420-480 g) underwent hHcy evoked by a Met-enriched diet (2 g/kg of weight/day) lasting four weeks. Changes in the metabolic ratio profile and significant histomorphological alterations have been found in the DG of hHcy rats. We have detected increased morphologically changed neurons and glial cells with increased neurogenic markers and apolipoprotein E positivity parallel with a diminished immunosignal for the N-Methyl-D-Aspartate receptor 1 in hHcy animals. A Met diet induced hHcy, likely via direct Hcy neurotoxicity, an interference with one carbon unit metabolism, and/or epigenetic regulation. These conditions lead to the progression of neurodegeneration and the promotion of AD-like pathological features in the less vulnerable hippocampal DG, which presents a plausible therapeutic target.
Collapse
Affiliation(s)
- Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Libusa Kovalska
- Clinic of Stomatology and Maxillofacial Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Jan Lehotsky
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
25
|
Robison LS, Gannon OJ, Salinero AE, Abi-Ghanem C, Kelly RD, Riccio DA, Mansour FM, Zuloaga KL. Sex differences in metabolic phenotype and hypothalamic inflammation in the 3xTg-AD mouse model of Alzheimer's disease. Biol Sex Differ 2023; 14:51. [PMID: 37559092 PMCID: PMC10410820 DOI: 10.1186/s13293-023-00536-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is notably associated with cognitive decline resulting from impaired function of hippocampal and cortical areas; however, several other domains and corresponding brain regions are affected. One such brain region is the hypothalamus, shown to atrophy and develop amyloid and tau pathology in AD patients. The hypothalamus controls several functions necessary for survival, including energy and glucose homeostasis. Changes in appetite and body weight are common in AD, often seen several years prior to the onset of cognitive symptoms. Therefore, altered metabolic processes may serve as a biomarker for AD, as well as a target for treatment, considering they are likely both a result of pathological changes and contributor to disease progression. Previously, we reported sexually dimorphic metabolic disturbances in ~ 7-month-old 3xTg-AD mice, accompanied by differences in systemic and hypothalamic inflammation. METHODS In the current study, we investigated metabolic outcomes and hypothalamic inflammation in 3xTg-AD males and females at 3, 6, 9, and 12 months of age to determine when these sex differences emerge. RESULTS In agreement with our previous study, AD males displayed less weight gain and adiposity, as well as reduced blood glucose levels following a glucose challenge, compared to females. These trends were apparent by 6-9 months of age, coinciding with increased expression of inflammatory markers (Iba1, GFAP, TNF-α, and IL-1β) in the hypothalamus of AD males. CONCLUSIONS These findings provide additional evidence for sex-dependent effects of AD pathology on energy and glucose homeostasis, which may be linked to hypothalamic inflammation.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL, 33328, USA.
| | - Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - David A Riccio
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
26
|
Damestani NL, Jacoby J, Yadav SM, Lovely AE, Michael A, Terpstra M, Eshghi M, Rashid B, Cruchaga C, Salat DH, Juttukonda MR. Associations between age, sex, APOE genotype, and regional vascular physiology in typically aging adults. Neuroimage 2023; 275:120167. [PMID: 37187365 PMCID: PMC10339339 DOI: 10.1016/j.neuroimage.2023.120167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
Altered blood flow in the human brain is characteristic of typical aging. However, numerous factors contribute to inter-individual variation in patterns of blood flow throughout the lifespan. To better understand the mechanisms behind such variation, we studied how sex and APOE genotype, a primary genetic risk factor for Alzheimer's disease (AD), influence associations between age and brain perfusion measures. We conducted a cross-sectional study of 562 participants from the Human Connectome Project - Aging (36 to >90 years of age). We found widespread associations between age and vascular parameters, where increasing age was associated with regional decreases in cerebral blood flow (CBF) and increases in arterial transit time (ATT). When grouped by sex and APOE genotype, interactions between group and age demonstrated that females had relatively greater CBF and lower ATT compared to males. Females carrying the APOEε4 allele showed the strongest association between CBF decline and ATT incline with age. This demonstrates that sex and genetic risk for AD modulate age-associated patterns of cerebral perfusion measures.
Collapse
Affiliation(s)
- Nikou L Damestani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA.
| | - John Jacoby
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Shrikanth M Yadav
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Allison E Lovely
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Aurea Michael
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Melissa Terpstra
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Barnaly Rashid
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
| | - David H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston MA, USA
| | - Meher R Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Berezin CT, Bergum N, Torres Lopez GM, Vigh J. Morphine pharmacokinetics and opioid transporter expression at the blood-retina barrier of male and female mice. Front Pharmacol 2023; 14:1206104. [PMID: 37388441 PMCID: PMC10301758 DOI: 10.3389/fphar.2023.1206104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Opioids are effective analgesics for treating moderate to severe pain, however, their use must be weighed against their dangerous side effects. Investigations into opioid pharmacokinetics provide crucial information regarding both on- and off-target drug effects. Our recent work showed that morphine deposits and accumulates in the mouse retina at higher concentrations than in the brain upon chronic systemic exposure. We also found reduced retinal expression of P-glycoprotein (P-gp), a major opioid extruder at the blood-brain barrier (BBB). Here, we systematically interrogated the expression of three putative opioid transporters at the blood-retina barrier (BRB): P-gp, breast cancer resistance protein (Bcrp) and multidrug resistance protein 2 (Mrp2). Using immunohistochemistry, we found robust expression of P-gp and Bcrp, but not Mrp2, at the inner BRB of the mouse retina. Previous studies have suggested that P-gp expression may be regulated by sex hormones. However, upon acute morphine treatment we found no sex differences in morphine deposition levels in the retina or brain, nor on transporter expression in the retinas of males and females with a high or low estrogen:progesterone ratio. Importantly, we found that P-gp, but not Bcrp, expression significantly correlated with morphine concentration in the retina, suggesting P-gp is the predominant opioid transporter at the BRB. In addition, fluorescence extravasation studies revealed that chronic morphine treatment did not alter the permeability of either the BBB or BRB. Together, these data suggest that reduced P-gp expression mediates retinal morphine accumulation upon systemic delivery, and in turn, potential effects on circadian photoentrainment.
Collapse
Affiliation(s)
- Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Glenda M. Torres Lopez
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jozsef Vigh
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
28
|
Easson K, Gilbert G, Gauthier C, Rohlicek CV, Saint-Martin C, Brossard-Racine M. Sex-Specific Cerebral Blood Flow Alterations in Youth Operated for Congenital Heart Disease. J Am Heart Assoc 2023:e028378. [PMID: 37301764 DOI: 10.1161/jaha.122.028378] [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: 11/03/2022] [Accepted: 03/23/2023] [Indexed: 06/12/2023]
Abstract
Background Lower cerebral blood flow (CBF) has previously been documented preoperatively in neonates with congenital heart disease (CHD). However, it remains unclear if these CBF deficits persist over the life span of CHD survivors following heart surgery. When exploring this question, it is critical to consider the sex differences in CBF that emerge during adolescence. Therefore, this study aimed to compare global and regional CBF between postpubertal youth with CHD and healthy peers and examine if such alterations are related to sex. Methods and Results Youth aged 16 to 24 years who underwent open heart surgery for complex CHD during infancy and age- and sex-matched controls completed brain magnetic resonance imaging, including T1-weighted and pseudo-continuous arterial spin labeling acquisitions. Global gray matter CBF and regional CBF in 9 bilateral gray matter regions were quantified for each participant. Compared with female controls (N=27), female participants with CHD (N=25) presented with lower global and regional CBF. In contrast, there were no differences in CBF between male controls (N=18) and males with CHD (N=17). Concurrently, female controls had higher global and regional CBF compared with male controls, with no differences in CBF between female and male participants with CHD. CBF was lower in individuals with a Fontan circulation. Conclusions This study provides evidence of altered CBF in postpubertal female participants with CHD despite undergoing surgical intervention during infancy. Alterations to CBF could have implications for later cognitive decline, neurodegeneration, and cerebrovascular disease in women with CHD.
Collapse
Affiliation(s)
- Kaitlyn Easson
- Advances in Brain & Child Development (ABCD) Research Laboratory Research Institute of the McGill University Health Centre Montreal Quebec Canada
- Department of Neurology & Neurosurgery, Faculty of Medicine & Health Sciences McGill University Quebec Montreal Canada
| | | | - Claudine Gauthier
- Department of Physics Concordia University Montreal Quebec Canada
- Montreal Heart Institute Research Centre Montreal Quebec Canada
| | - Charles V Rohlicek
- Division of Cardiology, Department of Pediatrics Montreal Children's Hospital Montreal Quebec Canada
| | - Christine Saint-Martin
- Division of Pediatric Radiology, Department of Medical Imaging Montreal Children's Hospital Montreal Quebec Canada
| | - Marie Brossard-Racine
- Advances in Brain & Child Development (ABCD) Research Laboratory Research Institute of the McGill University Health Centre Montreal Quebec Canada
- Department of Neurology & Neurosurgery, Faculty of Medicine & Health Sciences McGill University Quebec Montreal Canada
- Division of Neonatology, Department of Pediatrics Montreal Children's Hospital Montreal Quebec Canada
- School of Physical & Occupational Therapy, Faculty of Medicine and Health Sciences McGill University Quebec Montreal Canada
| |
Collapse
|
29
|
Sun F. The impact of blood pressure variability on cognition: current limitations and new advances. J Hypertens 2023; 41:888-905. [PMID: 37016905 PMCID: PMC10158606 DOI: 10.1097/hjh.0000000000003422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/18/2023] [Accepted: 02/23/2023] [Indexed: 04/06/2023]
Abstract
Dementia is the most common neurodegenerative disease in the aging population. Emerging evidence indicates that blood pressure (BP) variability is correlated with cognitive impairment and dementia independent of mean BP levels. The state-of-the-art review summarizes the latest evidence regarding the impact of BP variability on cognition in cognitively intact populations, patients with mild cognitive impairment, and different dementia types, focusing on the important confounding factors and new advances. This review also summarizes the potential mechanisms underlying the relationship between BP variability and cognitive impairment, and dementia, briefly discussing sex differences in the relationship. At last, current limitations and future perspectives are discussed to optimize BP management in preventing cognitive impairment and dementia.
Collapse
Affiliation(s)
- Fen Sun
- Department of Anatomy, College of Basic Medicine
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Gannon OJ, Naik JS, Riccio D, Mansour FM, Abi-Ghanem C, Salinero AE, Kelly RD, Brooks HL, Zuloaga KL. Menopause causes metabolic and cognitive impairments in a chronic cerebral hypoperfusion model of vascular contributions to cognitive impairment and dementia. Biol Sex Differ 2023; 14:34. [PMID: 37221553 DOI: 10.1186/s13293-023-00518-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/08/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND The vast majority of women with dementia are post-menopausal. Despite clinical relevance, menopause is underrepresented in rodent models of dementia. Before menopause, women are less likely than men to experience strokes, obesity, and diabetes-known risk factors for vascular contributions to cognitive impairment and dementia (VCID). During menopause, ovarian estrogen production stops and the risk of developing these dementia risk factors spikes. Here, we aimed to determine if menopause worsens cognitive impairment in VCID. We hypothesized that menopause would cause metabolic dysfunction and increase cognitive impairment in a mouse model of VCID. METHODS We performed a unilateral common carotid artery occlusion surgery to produce chronic cerebral hypoperfusion and model VCID in mice. We used 4-vinylcyclohexene diepoxide to induce accelerated ovarian failure and model menopause. We evaluated cognitive impairment using behavioral tests including novel object recognition, Barnes maze, and nest building. To assess metabolic changes, we measured weight, adiposity, and glucose tolerance. We explored multiple aspects of brain pathology including cerebral hypoperfusion and white matter changes (commonly observed in VCID) as well as changes to estrogen receptor expression (which may mediate altered sensitivity to VCID pathology post-menopause). RESULTS Menopause increased weight gain, glucose intolerance, and visceral adiposity. VCID caused deficits in spatial memory regardless of menopausal status. Post-menopausal VCID specifically led to additional deficits in episodic-like memory and activities of daily living. Menopause did not alter resting cerebral blood flow on the cortical surface (assessed by laser speckle contrast imaging). In the white matter, menopause decreased myelin basic protein gene expression in the corpus callosum but did not lead to overt white matter damage (assessed by Luxol fast blue). Menopause did not significantly alter estrogen receptor expression (ERα, ERβ, or GPER1) in the cortex or hippocampus. CONCLUSIONS Overall, we have found that the accelerated ovarian failure model of menopause caused metabolic impairment and cognitive deficits in a mouse model of VCID. Further studies are needed to identify the underlying mechanism. Importantly, the post-menopausal brain still expressed estrogen receptors at normal (pre-menopausal) levels. This is encouraging for any future studies attempting to reverse the effects of estrogen loss by activating brain estrogen receptors.
Collapse
Affiliation(s)
- Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Janvie S Naik
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
31
|
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: 8] [Impact Index Per Article: 4.0] [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
|
32
|
Prossnitz ER, Barton M. The G protein-coupled oestrogen receptor GPER in health and disease: an update. Nat Rev Endocrinol 2023:10.1038/s41574-023-00822-7. [PMID: 37193881 DOI: 10.1038/s41574-023-00822-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 05/18/2023]
Abstract
Oestrogens and their receptors contribute broadly to physiology and diseases. In premenopausal women, endogenous oestrogens protect against cardiovascular, metabolic and neurological diseases and are involved in hormone-sensitive cancers such as breast cancer. Oestrogens and oestrogen mimetics mediate their effects via the cytosolic and nuclear receptors oestrogen receptor-α (ERα) and oestrogen receptor-β (ERβ) and membrane subpopulations as well as the 7-transmembrane G protein-coupled oestrogen receptor (GPER). GPER, which dates back more than 450 million years in evolution, mediates both rapid signalling and transcriptional regulation. Oestrogen mimetics (such as phytooestrogens and xenooestrogens including endocrine disruptors) and licensed drugs such as selective oestrogen receptor modulators (SERMs) and downregulators (SERDs) also modulate oestrogen receptor activity in both health and disease. Following up on our previous Review of 2011, we herein summarize the progress made in the field of GPER research over the past decade. We will review molecular, cellular and pharmacological aspects of GPER signalling and function, its contribution to physiology, health and disease, and the potential of GPER to serve as a therapeutic target and prognostic indicator of numerous diseases. We also discuss the first clinical trial evaluating a GPER-selective drug and the opportunity of repurposing licensed drugs for the targeting of GPER in clinical medicine.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
- Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
33
|
Yoon CW, Rha JH, Park HK, Park SH, Kwon S, Kim BC, Youn YC, Jeong JH, Han HJ, Choi SH. Sex differences in the progression of cerebral microbleeds in patients with concomitant cerebral small vessel disease. Front Neurol 2022; 13:1054624. [PMID: 36619919 PMCID: PMC9810543 DOI: 10.3389/fneur.2022.1054624] [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: 09/27/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background and purpose Sex differences in cerebral microbleeds (CMBs) are not well-known. We aimed to assess the impact of sex on the progression of CMBs. Methods The CHALLENGE (Comparison Study of Cilostazol and Aspirin on Changes in Volume of Cerebral Small Vessel Disease White Matter Changes) database was analyzed. Out of 256 subjects, 189 participants with a follow-up brain scan were included in the analysis. The linear mixed-effect model was used to compare the 2-year changes in the number of CMBs between men and women. Results A total of 65 men and 124 women were analyzed. There were no significant differences in the prevalence (70.8 vs. 71.8%; P = 1.000) and the median [interquartile range (IQR)] number of total CMBs [1 (0-7) vs. 2 (0-7); P = 0.810] at baseline between men and women. The median (IQR) increase over 2 years in the number of CMBs was statistically higher in women than in men [1 (0-2) vs. 0 (0-1), P = 0.026]. The multivariate linear mixed-effects model showed that women had a significantly greater increase in the number of total, deep, and lobar CMBs compared to men after adjusting for age and the baseline number of CMBs [estimated log-transformed mean of difference between women and men: 0.040 (P = 0.028) for total CMBs, 0.037 (P = 0.047) for deep CMBs, and 0.047 (P = 0.009) for lobar CMBs]. Conclusion The progression of CMB over 2 years was significantly greater in women than in men.
Collapse
Affiliation(s)
- Cindy W. Yoon
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Joung-Ho Rha
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hee-Kwon Park
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Soo-Hyun Park
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Soonwook Kwon
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Hyun Jeong Han
- Department of Neurology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea,*Correspondence: Seong Hye Choi
| |
Collapse
|
34
|
Wang S, Tang C, Liu Y, Border JJ, Roman RJ, Fan F. Impact of impaired cerebral blood flow autoregulation on cognitive impairment. FRONTIERS IN AGING 2022; 3:1077302. [PMID: 36531742 PMCID: PMC9755178 DOI: 10.3389/fragi.2022.1077302] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/23/2022] [Indexed: 11/16/2023]
Abstract
Although the causes of cognitive impairment are multifactorial, emerging evidence indicates that cerebrovascular dysfunction plays an essential role in dementia. One of the most critical aspects of cerebrovascular dysfunction is autoregulation of cerebral blood flow (CBF), mainly mediated by the myogenic response, which is often impaired in dementia individuals with comorbidities, such as diabetes and hypertension. However, many unsolved questions remain. How do cerebrovascular networks coordinately modulate CBF autoregulation in health and disease? Does poor CBF autoregulation have an impact on cognitive impairment, and what are the underlying mechanisms? This review summarizes the cerebral vascular structure and myogenic (a three-phase model), metabolic (O2, CO2, adenosine, and H+), and endothelial (shear stress) factors in the regulation of CBF; and the consequences of CBF dysautoregulation. Other factors contributing to cerebrovascular dysfunction, such as impaired functional hyperemia and capillary abnormalities, are included as well. Moreover, this review highlights recent studies from our lab in terms of novel mechanisms involved in CBF autoregulation and addresses a hypothesis that there is a three-line of defense for CBF autoregulation in the cerebral vasculature.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Chengyun Tang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
35
|
Nuthikattu S, Milenkovic D, Norman JE, Rutledge J, Villablanca A. High Glycemia and Soluble Epoxide Hydrolase in Females: Differential Multiomics in Murine Brain Microvasculature. Int J Mol Sci 2022; 23:13044. [PMID: 36361847 PMCID: PMC9655872 DOI: 10.3390/ijms232113044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 12/02/2023] Open
Abstract
The effect of a high glycemic diet (HGD) on brain microvasculature is a crucial, yet understudied research topic, especially in females. This study aimed to determine the transcriptomic changes in female brain hippocampal microvasculature induced by a HGD and characterize the response to a soluble epoxide hydrolase inhibitor (sEHI) as a mechanism for increased epoxyeicosatrienoic acids (EETs) levels shown to be protective in prior models of brain injury. We fed mice a HGD or a low glycemic diet (LGD), with/without the sEHI (t-AUCB), for 12 weeks. Using microarray, we assessed differentially expressed protein-coding and noncoding genes, functional pathways, and transcription factors from laser-captured hippocampal microvessels. We demonstrated for the first time in females that the HGD had an opposite gene expression profile compared to the LGD and differentially expressed 506 genes, primarily downregulated, with functions related to cell signaling, cell adhesion, cellular metabolism, and neurodegenerative diseases. The sEHI modified the transcriptome of female mice consuming the LGD more than the HGD by modulating genes involved in metabolic pathways that synthesize neuroprotective EETs and associated with a higher EETs/dihydroxyeicosatrienoic acids (DHETs) ratio. Our findings have implications for sEHIs as promising therapeutic targets for the microvascular dysfunction that accompanies vascular dementia.
Collapse
Affiliation(s)
| | - Dragan Milenkovic
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Jennifer E. Norman
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA
| | - John Rutledge
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA
| | - Amparo Villablanca
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
36
|
Adesse D, Gladulich L, Alvarez-Rosa L, Siqueira M, Marcos AC, Heider M, Motta CS, Torices S, Toborek M, Stipursky J. Role of aging in Blood-Brain Barrier dysfunction and susceptibility to SARS-CoV-2 infection: impacts on neurological symptoms of COVID-19. Fluids Barriers CNS 2022; 19:63. [PMID: 35982454 PMCID: PMC9386676 DOI: 10.1186/s12987-022-00357-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/18/2022] [Indexed: 12/21/2022] Open
Abstract
COVID-19, which is caused by Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), has resulted in devastating morbidity and mortality worldwide due to lethal pneumonia and respiratory distress. In addition, the central nervous system (CNS) is well documented to be a target of SARS-CoV-2, and studies detected SARS-CoV-2 in the brain and the cerebrospinal fluid of COVID-19 patients. The blood-brain barrier (BBB) was suggested to be the major route of SARS-CoV-2 infection of the brain. Functionally, the BBB is created by an interactome between endothelial cells, pericytes, astrocytes, microglia, and neurons, which form the neurovascular units (NVU). However, at present, the interactions of SARS-CoV-2 with the NVU and the outcomes of this process are largely unknown. Moreover, age was described as one of the most prominent risk factors for hospitalization and deaths, along with other comorbidities such as diabetes and co-infections. This review will discuss the impact of SARS-CoV-2 on the NVU, the expression profile of SARS-CoV-2 receptors in the different cell types of the CNS and the possible role of aging in the neurological outcomes of COVID-19. A special emphasis will be placed on mitochondrial functions because dysfunctional mitochondria are also a strong inducer of inflammatory reactions and the "cytokine storm" associated with SARS-CoV-2 infection. Finally, we will discuss possible drug therapies to treat neural endothelial function in aged patients, and, thus, alleviate the neurological symptoms associated with COVID-19.
Collapse
Affiliation(s)
- Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil.
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Luis Gladulich
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Liandra Alvarez-Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michele Siqueira
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anne Caroline Marcos
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Marialice Heider
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Caroline Soares Motta
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Joice Stipursky
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
37
|
Subramaniapillai S, Suri S, Barth C, Maximov II, Voldsbekk I, van der Meer D, Gurholt TP, Beck D, Draganski B, Andreassen OA, Ebmeier KP, Westlye LT, de Lange AG. Sex- and age-specific associations between cardiometabolic risk and white matter brain age in the UK Biobank cohort. Hum Brain Mapp 2022; 43:3759-3774. [PMID: 35460147 PMCID: PMC9294301 DOI: 10.1002/hbm.25882] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/24/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiometabolic risk (CMR) factors are associated with accelerated brain aging and increased risk for sex-dimorphic illnesses such as Alzheimer's disease (AD). Yet, it is unknown how CMRs interact with sex and apolipoprotein E-ϵ4 (APOE4), a known genetic risk factor for AD, to influence brain age across different life stages. Using age prediction based on multi-shell diffusion-weighted imaging data in 21,308 UK Biobank participants, we investigated whether associations between white matter Brain Age Gap (BAG) and body mass index (BMI), waist-to-hip ratio (WHR), body fat percentage (BF%), and APOE4 status varied (i) between males and females, (ii) according to age at menopause in females, and (iii) across different age groups in males and females. We report sex differences in associations between BAG and all three CMRs, with stronger positive associations among males compared to females. Independent of APOE4 status, higher BAG (older brain age relative to chronological age) was associated with greater BMI, WHR, and BF% in males, whereas in females, higher BAG was associated with greater WHR, but not BMI and BF%. These divergent associations were most prominent within the oldest group of females (66-81 years), where greater BF% was linked to lower BAG. Earlier menopause transition was associated with higher BAG, but no interactions were found with CMRs. In conclusion, the findings point to sex- and age-specific associations between CMRs and brain age. Incorporating sex as a factor of interest in studies addressing CMR may promote sex-specific precision medicine, consequently improving health care for both males and females.
Collapse
Affiliation(s)
- Sivaniya Subramaniapillai
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of Psychology, Faculty of ScienceMcGill UniversityMontrealQuebecCanada
- Department of PsychologyUniversity of OsloOsloNorway
| | - Sana Suri
- Department of PsychiatryUniversity of OxfordOxfordUK
- Wellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
| | - Ivan I. Maximov
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
| | - Irene Voldsbekk
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- School of Mental Health and Neuroscience, Faculty of Health Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Tiril P. Gurholt
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| | - Dani Beck
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
| | - Bogdan Draganski
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of NeurologyMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | | | - Lars T. Westlye
- Department of PsychologyUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | - Ann‐Marie G. de Lange
- LREN, Centre for Research in Neurosciences, Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of PsychologyUniversity of OsloOsloNorway
- Department of PsychiatryUniversity of OxfordOxfordUK
| |
Collapse
|
38
|
Cilhoroz BT, DeBlois JP, Lefferts WK, Keller AP, Pagan Lassalle P, Meyer ML, Stoner L, Heffernan KS. Exploration of cerebral hemodynamic pathways through which large artery function affects neurovascular coupling in young women. Front Cardiovasc Med 2022; 9:914439. [PMID: 36035945 PMCID: PMC9411931 DOI: 10.3389/fcvm.2022.914439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe interactions between large artery function and neurovascular coupling (NVC) are emerging as important contributors to cognitive health. Women are disproportionally affected by Alzheimer's disease and related dementia later in life. Understanding large artery correlates of NVC in young women may help with preservation of cognitive health with advancing age.PurposeTo explore the association between large artery function, NVC and cognitive performance in young women.MethodsVascular measurements were made in 61 women (21 ± 4 yrs) at rest and during a cognitive challenge (Stroop task). Transcranial Doppler was used to measure left middle cerebral artery (MCA) maximum velocity (Vmax), mean velocity (Vmean), and pulsatility index (PI). NVC was determined as MCA blood velocity reactivity to the Stroop task. Large artery function was determined using carotid-femoral pulse wave velocity (cfPWV) as a proxy measure of aortic stiffness and carotid ultrasound-derived measures of compliance and reactivity (diameter change to the Stroop task). Cognitive function was assessed separately using a computerized neurocognitive battery that included appraisal of response speed, executive function, information processing efficiency, memory, attention/concentration, and impulsivity.ResultsMCA Vmax reactivity was positively associated with executive function (β = 0.26, 95% CI 0.01–0.10); MCA Vmean reactivity was negatively associated with response speed (β = −0.33, 95% CI −0.19 to −0.02) and positively with memory score (β = 0.28, 95% CI 0.01–0.19). MCA PI reactivity was negatively associated with attention performance (β = −0.29, 95% CI −14.9 to −1.0). Path analyses identified significant paths (p < 0.05) between carotid compliance and carotid diameter reactivity to select domains of cognitive function through MCA reactivity.ConclusionsNVC was associated with cognitive function in young women. Carotid artery function assessed as carotid compliance and carotid reactivity may contribute to optimal NVC in young women through increased blood flow delivery and reduced blood flow pulsatility.
Collapse
Affiliation(s)
- Burak T. Cilhoroz
- Department of Exercise Science, Syracuse University, Syracuse, NY, United States
| | - Jacob P. DeBlois
- Department of Exercise Science, Syracuse University, Syracuse, NY, United States
| | - Wesley K. Lefferts
- Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Allison P. Keller
- Department of Exercise Science, Syracuse University, Syracuse, NY, United States
| | - Patricia Pagan Lassalle
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, NC, United States
| | - Michelle L. Meyer
- Department of Epidemiology, Gilling's School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Emergency Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lee Stoner
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, NC, United States
- Department of Epidemiology, Gilling's School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kevin S. Heffernan
- Department of Exercise Science, Syracuse University, Syracuse, NY, United States
- *Correspondence: Kevin S. Heffernan
| |
Collapse
|
39
|
Zhu D, Zhang M, He B, Wan Y, Wang L, Gao F. The role of sex and ovarian hormones in hippocampal damage and cognitive deficits induced by chronic exposure to hypobaric hypoxia. Front Neurosci 2022; 16:953417. [PMID: 36003965 PMCID: PMC9393425 DOI: 10.3389/fnins.2022.953417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose This study aims to investigate the role of sex and ovarian hormones in hippocampal damage and cognitive deficits and behavioral dysfunction in rats induced by chronic exposure to hypobaric hypoxia. Methods Six-week-old male and female SD rats were housed for 3 months either in a real altitude (4,250 m) environment as the model of chronic hypobaric-hypoxia (CHH) or in a plain as controls. The animal behavioral and hippocampal neurons at subcellular, molecular, and ultrastructural levels were characterized after CHH exposure. Results After 3 months of CHH exposure, (1) male CHH rats’ serum testosterone level was lower than male controls’ whereas female CHH rats’ serum estradiol level was higher than female controls’; (2) Morris water maze test finds that male rats showed more learning and spatial memory deficits than female rats; (3) male rats showed more severe hippocampal damage, hippocampal inflammation, oxidative stress and decreased hippocampal integrity (neurogenesis and dendritic spine density) than female rats; (4) Western blot analysis shows that, compared with the male control group, in male CHH group’s hippocampus, expression of nNOS, HO-1, and Bax protein increased whereas that of Bcl-2 protein decreased; (5) Expression of PON2 protein in male rats (CHH and controls) was lower than female rats (CHH and controls). In addition, CHH exposure decreased the expression of PON2 protein in both male and female rats; (6) qPCR analysis reveals that CHH exposure reduced the gene expression of N-methyl-D-aspartate receptor NR2A and NR2B subunits in male rats’ hippocampus. In addition, compared with the sham CHH group, the expression level of PON2 protein decreased in the OVX-CHH group’s hippocampus whereas oxidative stress, neuroinflammation, and degeneration of hippocampal neurons increased in the OVX-CHH group’s hippocampus. Conclusion After CHH exposure, male rats were significantly more likely than female rats to develop hippocampal damage, hippocampal neuroinflammation, and cognitive decline and deficits, suggesting that sex and ovarian hormones were significantly involved in regulating the rats’ susceptibility to CHH exposure-induced hippocampal damage.
Collapse
|
40
|
Salwierz P, Davenport C, Sumra V, Iulita MF, Ferretti MT, Tartaglia MC. Sex and gender differences in dementia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:179-233. [PMID: 36038204 DOI: 10.1016/bs.irn.2022.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The dementia landscape has undergone a striking paradigm shift. The advances in understanding of neurodegeneration and proteinopathies has changed our approach to patients with cognitive impairment. Firstly, it has recently been shown that the various proteinopathies that are the cause of the dementia begin to build up long before the appearance of any obvious symptoms. This has cemented the idea that there is an urgency in diagnosis as it occurs very late in the pathophysiology of these diseases. Secondly, that accurate diagnosis is required to deliver targeted therapies, that is precision medicine. With this latter point, the realization that various factors of a person need to be considered as they may impact the presentation and progression of disease has risen to the forefront. Two of these factors aside from race and age are biological sex and gender (social construct), as both can have tremendous impact on manifestation of disease. This chapter will cover what is known and remains to be known on the interaction of sex and gender with some of the major causes of dementia.
Collapse
Affiliation(s)
- Patrick Salwierz
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Carly Davenport
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Vishaal Sumra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - M Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Women's Brain Project, Guntershausen, Switzerland
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Memory Clinic, Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
41
|
Leacy JK, Johnson EM, Lavoie LR, Macilwraith DN, Bambury M, Martin JA, Lucking EF, Linares AM, Saran G, Sheehan DP, Sharma N, Day TA, O'Halloran KD. Variation within the visually evoked neurovascular coupling response of the posterior cerebral artery is not influenced by age or sex. J Appl Physiol (1985) 2022; 133:335-348. [PMID: 35771218 PMCID: PMC9359642 DOI: 10.1152/japplphysiol.00292.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurovascular coupling (NVC) is the temporal and spatial coordination between local neuronal activity and regional cerebral blood flow. The literature is unsettled on whether age and/or sex affect NVC, which may relate to differences in methodology and the quantification of NVC in small sample-sized studies. The aim of this study was to 1) determine the relative and combined contribution of age and sex to the variation observed across several distinct NVC metrics (n = 125, 21–66 yr; 41 males) and 2) present an approach for the comprehensive systematic assessment of the NVC response using transcranial Doppler ultrasound. NVC was measured as the relative change from baseline (absolute and percent change) assessing peak, mean, and total area under the curve (tAUC) of cerebral blood velocity through the posterior cerebral artery (PCAv) during intermittent photic stimulation. In addition, the NVC waveform was compartmentalized into distinct regions, acute (0–9 s), mid (10–19 s), and late (20–30 s), following the onset of photic stimulation. Hierarchical multiple regression modeling was used to determine the extent of variation within each NVC metric attributable to demographic differences in age and sex. After controlling for differences in baseline PCAv, the R2 data suggest that 1.6%, 6.1%, 1.1%, 3.4%, 2.5%, and 4.2% of the variance observed within mean, peak, tAUC, acute, mid, and late response magnitude is attributable to the combination of age and sex. Our study reveals that variability in NVC response magnitude is independent of age and sex in healthy human participants, aged 21–66 yr. NEW & NOTEWORTHY We assessed the variability within the neurovascular coupling response attributable to age and sex (n = 125, 21–66 yr; 41 male). Based on the assessment of posterior cerebral artery responses to visual stimulation, 0%–6% of the variance observed within several metrics of NVC response magnitude are attributable to the combination of age and sex. Therefore, observed differences between age groups and/or sexes are likely a result of other physiological factors.
Collapse
Affiliation(s)
- Jack K Leacy
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Emily M Johnson
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Lauren R Lavoie
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Diane N Macilwraith
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Megan Bambury
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Jason A Martin
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Andrea M Linares
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Gurkarn Saran
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Dwayne P Sheehan
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Nishan Sharma
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Trevor A Day
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, Alberta, Canada
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
42
|
Johnson AC, Uhlig F, Einwag Z, Cataldo N, Erdos B. The neuroendocrine stress response impairs hippocampal vascular function and memory in male and female rats. Neurobiol Dis 2022; 168:105717. [PMID: 35385769 PMCID: PMC9018625 DOI: 10.1016/j.nbd.2022.105717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Chronic psychological stress affects brain regions involved in memory such as the hippocampus and accelerates age-related cognitive decline, including in Alzheimer's disease and vascular dementia. However, little is known about how chronic stress impacts hippocampal vascular function that is critically involved in maintaining neurocognitive health that could contribute to stress-related memory dysfunction. Here, we used a novel experimental rat model that mimics the neuroendocrine and cardiovascular aspects of chronic stress to determine how the neuroendocrine components of the stress response affect hippocampal function. We studied both male and female rats to determine potential sex differences in the susceptibility of the hippocampus and its vasculature to neuroendocrine stress-induced dysfunction. We show that activation of neuroendocrine stress pathways impaired the vasoreactivity of hippocampal arterioles to mediators involved in coupling neuronal activity with local blood flow that was associated with impaired memory function. Interestingly, we found more hippocampal arteriolar dysfunction and scarcer hippocampal microvasculature in male compared to female rats that was associated with greater memory impairment, suggesting the male sex may be at increased risk of neuroendocrine-derived hippocampal dysfunction during chronic stress. Overall, this study revealed the therapeutic potential of targeting hippocampal arterioles to prevent or slow memory decline in the setting of prolonged and/or unavoidable stress.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Friederike Uhlig
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Zachary Einwag
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Noelle Cataldo
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
43
|
Abstract
Brain disease remains a significant health, social, and economic burden with a high failure rate of translation of therapeutics to the clinic. Nanotherapeutics have represented a promising area of technology investment to improve drug bioavailability and delivery to the brain, with several successes for nanotherapeutic use for central nervous system disease that are currently in the clinic. However, renewed and continued research on the treatment of neurological disorders is critically needed. We explore the challenges of drug delivery to the brain and the ways in which nanotherapeutics can overcome these challenges. We provide a summary and overview of general design principles that can be applied to nanotherapeutics for uptake and penetration in the brain. We next highlight remaining questions that limit the translational potential of nanotherapeutics for application in the clinic. Lastly, we provide recommendations for ongoing preclinical research to improve the overall success of nanotherapeutics against neurological disease.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA;
| |
Collapse
|
44
|
Impact of diabetes and ischemic stroke on the cerebrovasculature: A female perspective. Neurobiol Dis 2022; 167:105667. [PMID: 35227927 PMCID: PMC9615543 DOI: 10.1016/j.nbd.2022.105667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/25/2022] [Accepted: 02/17/2022] [Indexed: 01/16/2023] Open
Abstract
There is a very complex interaction between the brain and the cerebral vasculature to meet the metabolic demands of the brain for proper function. Preservation of vascular networks and cerebrovascular function ultimately plays a key role in this intricate communication within the brain in health and disease. Experimental evidence showed that diabetes not only affects the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression, but also alters cerebrovascular function resulting in compromised myogenic reactivity and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain can rapidly occur. When an ischemic insult is superimposed on this pathology, not only is the neurovascular injury greater, but repair mechanisms fail, resulting in greater physical and cognitive deficits. While clinically it is known that women suffer disproportionately from diabetes as well as ischemic stroke and post-stroke cognitive impairment, the cerebrovascular architecture, patho/physiology, as well as cerebrovascular contributions to stroke recovery in female and diabetic animal models are inadequately studied and highlighted in this review.
Collapse
|
45
|
Bachmann D, Roman ZJ, Buchmann A, Zuber I, Studer S, Saake A, Rauen K, Gruber E, Nitsch RM, Hock C, Gietl AF, Treyer V. Lifestyle affects amyloid burden and cognition differently in men and women. Ann Neurol 2022; 92:451-463. [PMID: 35598071 PMCID: PMC9542817 DOI: 10.1002/ana.26417] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022]
Abstract
Objective Evidence on associations of lifestyle factors with Alzheimer's pathology and cognition are ambiguous, potentially because they rarely addressed inter‐relationships of factors and sex effects. While considering these aspects, we examined the relationships of lifestyle factors with brain amyloid burden and cognition. Methods We studied 178 cognitively normal individuals (women, 49%; 65.0 [7.6] years) and 54 individuals with mild cognitive impairment (women, 35%; 71.3 [8.3] years) enrolled in a prospective study of volunteers who completed 18F‐Flutemetamol amyloid positron emission tomography. Using structural equation modeling, we examined associations between latent constructs representing metabolic/vascular risk, physical activity, and cognitive activity with global amyloid burden and cognitive performance. Furthermore, we investigated the influence of sex in this model. Results Overall, higher cognitive activity was associated with better cognitive performance and higher physical activity was associated with lower amyloid burden. The latter association was weakened to a nonsignificant level after excluding multivariate outliers. Examination of the moderating effect of sex in the model revealed an inverse association of metabolic/vascular risk with cognition in men, whereas in women metabolic/vascular risk trended toward increased amyloid burden. Furthermore, a significant inverse association between physical activity and amyloid burden was found only in men. Inheritance of an APOE4 allele was associated with higher amyloid burden only in women. Interpretation Sex modifies effects of certain lifestyle‐related factors on amyloid burden and cognition. Notably, our results suggest that the negative impact of metabolic/vascular risk influences the risk of cognitive decline and Alzheimer's disease through distinct paths in women and men. ANN NEUROL 2022;92:451–463
Collapse
Affiliation(s)
- Dario Bachmann
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Zachary J Roman
- Department of Psychology, Psychological Methods, Evaluation, and Statistics, University of Zurich, Zurich, Switzerland
| | - Andreas Buchmann
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Isabelle Zuber
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Sandro Studer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Antje Saake
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Katrin Rauen
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Department of Geriatric Psychiatry, Psychiatric Hospital Zurich
| | - Esmeralda Gruber
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Neurimmune, Schlieren, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Neurimmune, Schlieren, Zurich, Switzerland
| | - Anton F Gietl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Department of Geriatric Psychiatry, Psychiatric Hospital Zurich
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Gannon OJ, Robison LS, Salinero AE, Abi-Ghanem C, Mansour FM, Kelly RD, Tyagi A, Brawley RR, Ogg JD, Zuloaga KL. High-fat diet exacerbates cognitive decline in mouse models of Alzheimer's disease and mixed dementia in a sex-dependent manner. J Neuroinflammation 2022; 19:110. [PMID: 35568928 PMCID: PMC9107741 DOI: 10.1186/s12974-022-02466-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/21/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Approximately 70% of Alzheimer's disease (AD) patients have co-morbid vascular contributions to cognitive impairment and dementia (VCID); this highly prevalent overlap of dementia subtypes is known as mixed dementia (MxD). AD is more prevalent in women, while VCID is slightly more prevalent in men. Sex differences in risk factors may contribute to sex differences in dementia subtypes. Unlike metabolically healthy women, diabetic women are more likely to develop VCID than diabetic men. Prediabetes is 3× more prevalent than diabetes and is linked to earlier onset of dementia in women, but not men. How prediabetes influences underlying pathology and cognitive outcomes across different dementia subtypes is unknown. To fill this gap in knowledge, we investigated the impact of diet-induced prediabetes and biological sex on cognitive function and neuropathology in mouse models of AD and MxD. METHODS Male and female 3xTg-AD mice received a sham (AD model) or unilateral common carotid artery occlusion surgery to induce chronic cerebral hypoperfusion (MxD model). Mice were fed a control or high fat (HF; 60% fat) diet from 3 to 7 months of age. In both sexes, HF diet elicited a prediabetic phenotype (impaired glucose tolerance) and weight gain. RESULTS In females, but not males, metabolic consequences of a HF diet were more severe in AD or MxD mice compared to WT. In both sexes, HF-fed AD or MxD mice displayed deficits in spatial memory in the Morris water maze (MWM). In females, but not males, HF-fed AD and MxD mice also displayed impaired spatial learning in the MWM. In females, but not males, AD or MxD caused deficits in activities of daily living, regardless of diet. Astrogliosis was more severe in AD and MxD females compared to males. Further, AD/MxD females had more amyloid beta plaques and hippocampal levels of insoluble amyloid beta 40 and 42 than AD/MxD males. In females, but not males, more severe glucose intolerance (prediabetes) was correlated with increased hippocampal microgliosis. CONCLUSIONS High-fat diet had a wider array of metabolic, cognitive, and neuropathological consequences in AD and MxD females compared to males. These findings shed light on potential underlying mechanisms by which prediabetes may lead to earlier dementia onset in women.
Collapse
Affiliation(s)
- Olivia J. Gannon
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Lisa S. Robison
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA ,grid.261241.20000 0001 2168 8324Department of Psychology & Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314 USA ,grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Abigail E. Salinero
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Charly Abi-Ghanem
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Febronia M. Mansour
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Richard D. Kelly
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Alvira Tyagi
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| | - Rebekah R. Brawley
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Jordan D. Ogg
- grid.264307.40000 0000 9688 1551Department of Psychology, Stetson University, 421 N Woodland Blvd, DeLand, FL 32723 USA
| | - Kristen L. Zuloaga
- grid.413558.e0000 0001 0427 8745Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY 12208 USA
| |
Collapse
|
47
|
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: 2.3] [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
|
48
|
Bumgarner JR, Nelson RJ. Open-source analysis and visualization of segmented vasculature datasets with VesselVio. CELL REPORTS METHODS 2022; 2:100189. [PMID: 35497491 PMCID: PMC9046271 DOI: 10.1016/j.crmeth.2022.100189] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/10/2022] [Accepted: 03/02/2022] [Indexed: 05/11/2023]
Abstract
Vascular networks are fundamental components of biological systems. Quantitative analysis and observation of the features of these networks can improve our understanding of their roles in health and disease. Recent advancements in imaging technologies have enabled the generation of large-scale vasculature datasets, but barriers to analyzing these datasets remain. Modern analysis options are mainly limited to paid applications or open-source terminal-based software that requires programming knowledge with high learning curves. Here, we describe VesselVio, an open-source application developed to analyze and visualize pre-binarized vasculature datasets and pre-constructed vascular graphs. Vasculature datasets and graphs can be loaded with annotations and processed with custom parameters. Here, the program is tested on ground-truth datasets and is compared with current pipelines. The utility of VesselVio is demonstrated by the analysis of multiple formats of 2D and 3D datasets acquired with several imaging modalities, including annotated mouse whole-brain vasculature volumes.
Collapse
Affiliation(s)
- Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
49
|
Wareham LK, Liddelow SA, Temple S, Benowitz LI, Di Polo A, Wellington C, Goldberg JL, He Z, Duan X, Bu G, Davis AA, Shekhar K, Torre AL, Chan DC, Canto-Soler MV, Flanagan JG, Subramanian P, Rossi S, Brunner T, Bovenkamp DE, Calkins DJ. Solving neurodegeneration: common mechanisms and strategies for new treatments. Mol Neurodegener 2022; 17:23. [PMID: 35313950 PMCID: PMC8935795 DOI: 10.1186/s13024-022-00524-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Across neurodegenerative diseases, common mechanisms may reveal novel therapeutic targets based on neuronal protection, repair, or regeneration, independent of etiology or site of disease pathology. To address these mechanisms and discuss emerging treatments, in April, 2021, Glaucoma Research Foundation, BrightFocus Foundation, and the Melza M. and Frank Theodore Barr Foundation collaborated to bring together key opinion leaders and experts in the field of neurodegenerative disease for a virtual meeting titled "Solving Neurodegeneration". This "think-tank" style meeting focused on uncovering common mechanistic roots of neurodegenerative disease and promising targets for new treatments, catalyzed by the goal of finding new treatments for glaucoma, the world's leading cause of irreversible blindness and the common interest of the three hosting foundations. Glaucoma, which causes vision loss through degeneration of the optic nerve, likely shares early cellular and molecular events with other neurodegenerative diseases of the central nervous system. Here we discuss major areas of mechanistic overlap between neurodegenerative diseases of the central nervous system: neuroinflammation, bioenergetics and metabolism, genetic contributions, and neurovascular interactions. We summarize important discussion points with emphasis on the research areas that are most innovative and promising in the treatment of neurodegeneration yet require further development. The research that is highlighted provides unique opportunities for collaboration that will lead to efforts in preventing neurodegeneration and ultimately vision loss.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Sally Temple
- Neural Stem Cell Institute, NY, 12144, Rensselaer, USA
| | - Larry I Benowitz
- Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, CA, Palo Alto, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, MA, Boston, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Albert A Davis
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, CA, 91125, Pasadena, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Aurora, CO, USA
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | | | | | | | | | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
50
|
Palmer JA, Kaufman CS, Vidoni ED, Honea RA, Burns JM, Billinger SA. Sex Differences in Resilience and Resistance to Brain Pathology and Dysfunction Moderated by Cerebrovascular Response to Exercise and Genetic Risk for Alzheimer's Disease. J Alzheimers Dis 2022; 90:535-542. [PMID: 36155505 PMCID: PMC9731318 DOI: 10.3233/jad-220359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Sex as a biological variable appears to contribute to the multifactorial etiology of Alzheimer's disease. We tested sex-based interactions between cerebrovascular function and APOE4 genotype on resistance and resilience to brain pathology and cognitive executive dysfunction in cognitively-normal older adults. Female APOE4 carriers had higher amyloid-β deposition yet achieved similar cognitive performance to males and female noncarriers. Further, female APOE4 carriers with robust cerebrovascular responses to exercise possessed lower amyloid-β. These results suggest a unique cognitive resilience and identify cerebrovascular function as a key mechanism for resistance to age-related brain pathology in females with high genetic vulnerability to Alzheimer's disease.
Collapse
Affiliation(s)
- Jacqueline A. Palmer
- Department of Neurology, School of Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America,University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Carolyn S. Kaufman
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Robyn A. Honea
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Sandra A. Billinger
- Department of Neurology, School of Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America,University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America,Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA,Correspondence: Sandra A. Billinger, PT, PhD, FAHA, , Twitter: @Sandy_REACHlab
| |
Collapse
|