1
|
Schreiber S, Arndt P, Morton L, Garza AP, Müller P, Neumann K, Mattern H, Dörner M, Bernal J, Vielhaber S, Meuth SG, Dunay IR, Dityatev A, Henneicke S. Immune system activation and cognitive impairment in arterial hypertension. Am J Physiol Cell Physiol 2024; 327:C1577-C1590. [PMID: 39495252 DOI: 10.1152/ajpcell.00219.2024] [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: 04/04/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Chronic arterial hypertension disrupts the integrity of the cerebral microvasculature, doubling the risk of age-related dementia. Despite sufficient antihypertensive therapy in still a significant proportion of individuals blood pressure lowering alone does not preserve cognitive health. Accumulating evidence highlights the role of inflammatory mechanisms in the pathogenesis of hypertension. In this review, we introduce a temporal framework to explore how early immune system activation and interactions at neurovascular-immune interfaces pave the way to cognitive impairment. The overall paradigm suggests that prohypertensive stimuli induce mechanical stress and systemic inflammatory responses that shift peripheral and meningeal immune effector mechanisms toward a proinflammatory state. Neurovascular-immune interfaces in the brain include a dysfunctional blood-brain barrier, crossed by peripheral immune cells; the perivascular space, in which macrophages respond to cerebrospinal fluid- and blood-derived immune regulators; and the meningeal immune reservoir, particularly T cells. Immune responses at these interfaces bridge peripheral and neurovascular unit inflammation, directly contributing to impaired brain perfusion, clearance of toxic metabolites, and synaptic function. We propose that deep immunophenotyping in biofluids together with advanced neuroimaging could aid in the translational determination of sequential immune and brain endotypes specific to arterial hypertension. This could close knowledge gaps on how and when immune system activation transits into neurovascular dysfunction and cognitive impairment. In the future, targeting specific immune mechanisms could prevent and halt hypertension disease progression before clinical symptoms arise, addressing the need for new interventions against one of the leading threats to cognitive health.
Collapse
Affiliation(s)
- Stefanie Schreiber
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Arndt
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
| | - Lorena Morton
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Patrick Müller
- Department of Cardiology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Katja Neumann
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Biomedical Magnetic Resonance, Faculty of Natural Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Marc Dörner
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Switzerland
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ildiko R Dunay
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Solveig Henneicke
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
| |
Collapse
|
2
|
Song Y, Zhou X, Zhao H, Zhao W, Sun Z, Zhu J, Yu Y. Characterizing the role of the microbiota-gut-brain axis in cerebral small vessel disease: An integrative multi‑omics study. Neuroimage 2024; 303:120918. [PMID: 39505226 DOI: 10.1016/j.neuroimage.2024.120918] [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: 04/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Prior efforts have revealed changes in gut microbiome, circulating metabolome, and multimodal neuroimaging features in cerebral small vessel disease (CSVD). However, there is a paucity of research integrating the multi-omic information to characterize the role of the microbiota-gut-brain axis in CSVD. METHODS We collected gut microbiome, fecal and blood metabolome, multimodal magnetic resonance imaging data from 37 CSVD patients with white matter hyperintensities and 46 healthy controls. Between-group comparison was performed to identify the differential gut microbial taxa, followed by performance of multi-stage microbiome-metabolome-neuroimaging-neuropsychology correlation analyses in CSVD patients. RESULTS Our data showed both depleted and enriched gut microbes in CSVD patients. Among the differential microbes, Haemophilus and Akkermansia were associated with a range of metabolites enriched for Aminoacyl-tRNA biosynthesis pathway. Furthermore, the affected metabolites were associated with neuroimaging measures involving gray matter morphology, spontaneous intrinsic brain activity, white matter integrity, and global structural network topology, which were in turn related to cognition and emotion in CSVD patients. CONCLUSION Our findings provide an integrative framework to understand the pathophysiological mechanisms underlying the interplay between gut microbiota dysbiosis and CSVD, highlighting the potential of targeting the microbiota-gut-brain axis as a therapeutic strategy in CSVD patients.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Han Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Wenming Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China.
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China.
| |
Collapse
|
3
|
Luo BL, He SP, Zhang YF, Yang QW, Zhuang JC, Zhu RJ, Zheng YQ, Su HM. Correlation between matrix metalloproteinase-2, matrix metalloproteinase-9, and tissue inhibitor of metalloproteinases-1 and white matter hyperintensities in patients with cerebral small vessel disease based on cranial magnetic resonance 3D imaging. Magn Reson Imaging 2024; 113:110213. [PMID: 39053592 DOI: 10.1016/j.mri.2024.110213] [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: 05/07/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE The objective of this study was to investigate the correlation between serum levels of matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), and tissue inhibitor of metalloproteinases-1 (TIMP-1) levels and their ratios with the severity of white matter hyperintensities (WMHs) in patients with cerebral small vessel disease (CSVD). METHODS This cross-sectional study was done on a prospective cohort of patients with CSVD. Qualitative and quantitative analyses of WMHs were performed using Fazekas grading and lesion prediction algorithm (LPA) methods. Biomarkers MMP-2, MMP-9, and TIMP-1 were measured to explore their correlation with the severity of WMHs. RESULTS The sample consisted of 144 patients with CSVD. There were 63 male and 81 female patients, with an average age of 67.604 ± 8.727 years. Among these, 58.33% presented with white matter hyperintensities at Fazekas grading level 1, with an average total template volume of WMHs of 4.305 mL. MMP-2 (P = 0.025), MMP-9 (P = 0.008), TIMP-1 (P = 0.026), and age (P = 0.007) were identified as independent correlates of WMHs based on Fazekas grading. Independent correlates of the total template volume of WMHs included MMP-2 (P = 0.023), TIMP-1 (P = 0.046), age (P = 0.047), systolic blood pressure (P = 0.047), and homocysteine (Hcy) (P = 0.014). In addition, age (P = 0.003; P < 0.001), interleukin-6 (IL-6) (P < 0.001; P = 0.044), Hcy (P < 0.001; P < 0.001), glycated hemoglobin (HbA1c) (P = 0.016; P = 0.043), and chronic kidney disease (P < 0.001; P < 0.001) were associated with both WMHs Fazekas grading and the total template volume of WMHs. CONCLUSION Serum levels of MMP-9, MMP-2, and TIMP-1 were independently associated with the Fazekas grading, while serum TIMP-1 and MMP-2 levels were independently related to the total template volume of WMHs. The association of TIMP-1 and MMP-2 with the severity of CSVD-related WMHs suggests their potential role as disease-related biomarkers. However, further research is required to uncover the specific mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Bei-Lin Luo
- The Graduate School of Fujian Medical University, Fuzhou 350000, China; Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| | - Shun-Po He
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| | - Yi-Fen Zhang
- The Graduate School of Fujian Medical University, Fuzhou 350000, China; Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| | - Qing-Wei Yang
- The Graduate School of Fujian Medical University, Fuzhou 350000, China; Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China.
| | - Jing-Cong Zhuang
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China.
| | - Ren-Jing Zhu
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| | - Ya-Qin Zheng
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| | - Hua-Mei Su
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, Fujian, China
| |
Collapse
|
4
|
Seidel F, Vreeken D, Custers E, Wiesmann M, Özsezen S, van Duyvenvoorde W, Caspers M, Menke A, Morrison MC, Verschuren L, Duering M, Hazebroek EJ, Kiliaan AJ, Kleemann R. Metabolic dysfunction-associated steatotic liver disease is associated with effects on cerebral perfusion and white matter integrity. Heliyon 2024; 10:e38516. [PMID: 39391513 PMCID: PMC11466594 DOI: 10.1016/j.heliyon.2024.e38516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
It is unclear whether early metabolic and inflammatory aberrations in the liver are associated with detrimental changes in brain structure and cognitive function. This cross-sectional study examines putative associations between metabolic dysfunction-associated steatotic liver disease (MASLD) and brain health in 36-55 year-old participants with obesity (n = 70) from the BARICO study (BAriatric surgery Rijnstate and Radboudumc neuroImaging and Cognition in Obesity). The participants underwent brain magnetic resonance imaging to study brain volumes and cortical thickness (3T MRI including T1-weighted magnetization-prepared rapid gradient-echo sequence), cerebral blood perfusion (arterial spin labeling) and white matter integrity (diffusion weighted imaging to assess mean-skeletonized mean diffusivity and fluid-attenuated inversion recovery to detect the presence of white matter hyperintensities (WMH)). The participants additionally performed neuropsychological tests to assess global cognition, working and episodic memory, verbal fluency and the ability to shift attention. Liver biopsies were collected and liver dysfunction was examined with histopathological, biochemical, and gene expression analyses. Linear regression analyses were performed between liver and brain parameters and the influence of body-mass index, diabetes and hypertension was explored. Early stages of liver disease were not associated with cognitive status but with cerebrovascular changes independently of age, sex, BMI, diabetes and hypertension: hepatic fibrosis development was associated with higher spatial coefficient of variation (sCoV) in the nucleus accumbens (NAcc), reflecting greater variations in cerebral perfusion and reduced vascular efficiency. Elevated hepatic levels of free cholesterol and cholesteryl esters were associated with increased WMH, indicating cerebral small vessel disease. RNA-seq and pathway analyses identified associations between sCoV in NAcc and WMH and the expression of hepatic genes involved in inflammation and cellular stress. Additionally, sCoV in NAcc correlated with plasma IL-6 levels suggesting that systemic-low grade inflammation may, at least partly, mediate this relationship. In conclusion, this study demonstrates that specific features of liver dysfunction (e.g. free cholesterol, onset of fibrosis) are associated with subtle cerebrovascular impairments, when changes in cognitive performance are not yet noticeable. These findings highlight the need for future research on therapeutic strategies that normalize metabolic-inflammatory aberrations in the liver to reduce the risk of cognitive decline.
Collapse
Affiliation(s)
- Florine Seidel
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Debby Vreeken
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
| | - Emma Custers
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
| | - Maximilian Wiesmann
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
| | - Serdar Özsezen
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Martien Caspers
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany
- Medical Imaging Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Marktgasse 8, CH-4051 Basel, Switzerland
| | - Eric J. Hazebroek
- Department of Bariatric Surgery, Vitalys, part of Rijnstate hospital, Postbus 9555, 6800 TA Arnhem, the Netherlands
- Division of Human Nutrition and Health, Wageningen University, Postbus 17 6700 AA Wageningen Wageningen, the Netherlands
| | - Amanda J. Kiliaan
- Department Medical Imaging, Anatomy, Radboud Alzheimer Center, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE Leiden, the Netherlands
| |
Collapse
|
5
|
Brett BL, Klein A, Vazirnia P, Omidfar S, Guskiewicz K, McCrea MA, Meier TB. White Matter Hyperintensities and Microstructural Alterations in Contact Sport Athletes from Adolescence to Early Midlife. J Neurotrauma 2024; 41:2307-2322. [PMID: 38661548 PMCID: PMC11564850 DOI: 10.1089/neu.2023.0609] [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: 04/26/2024] Open
Abstract
Studies have demonstrated associations between cumulative concussion and repetitive head impact exposure (RHI) through contact sports with white matter (WM) alterations later in life. The course of WM changes associated with exposure earlier in the lifespan is unclear. This study investigated alterations in white matter (WM hyperintensity [WMH] volume and microstructural changes) associated with concussion and RHI exposure from adolescence to early midlife, as well as the interaction between exposure and age cohort (i.e., adolescent/young adult compared with early midlife athlete cohorts) on WM outcomes. Participating football players included an adolescent/young adulthood cohort (n = 82; Mage = 18.4 ± 1.7) and an early midlife cohort (37 former collegiate players approximately 15 years removed from sport; Mage = 37.7 ± 1.4). Years of football participation and number of prior concussions were exposures of interest. White matter outcomes included log-transformed manually segmented total WMH volume and neurite orientation dispersion and density imaging metrics of microstructure/organization (isotropic volume fraction [Viso], intracellular volume fraction [Vic], and orientation dispersion [OD]). Regression models were fit to test the effects of concussion history, years of football participation, and age cohort by years of football participation with WM outcomes. Spearman's correlations assessed associations between significant WM metrics and measures of cognitive and psychological function. A significant age cohort by years of participation effect was observed for whole brain white matter OD, B = -0.002, SE = 0.001, p = 0.001. The interaction was driven by a negative association between years of participation and OD within the younger cohort, B = -0.001, SE = 0.0004, p = 0.008, whereas a positive association between participation and OD in the early midlife cohort, B = 0.001, SE = 0.0003, p = 0.039, was observed. Follow-up ROI analyses showed significant interaction effects for OD in the body of the corpus callosum, genu of the corpus callosum, cingulum, inferior fronto-occipital fasciculus, superior longitudinal fasciculus, and posterior thalamic radiation (p values <0.05). Greater concussion history was significantly associated with greater Viso in the early midlife cohort, B = 0.001, SE = 0.0002, p = 0.010. Years of participation and concussion history were not associated with WMH volume, p values >0.05. Performance on a measure of executive function was significantly associated with years of participation, ρ = 0.34, p = 0.04, and a trend was observed for OD, ρ = 0.28, p = 0.09 in the early midlife cohort only. The global characterization of white matter changes associated with years of football participation were broadly similar and stable from adolescence through early midlife (i.e., microstructural alterations, but not macroscopic lesions). An inverse association between years of participation and orientation dispersion across age cohorts may represent a process of initial recovery/reorganization proximal to sport, followed by later reduction of white matter coherence.
Collapse
Affiliation(s)
- Benjamin L. Brett
- Departments of Neurosurgery and Neurology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andrew Klein
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Parsia Vazirnia
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Samantha Omidfar
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kevin Guskiewicz
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
6
|
Zhao X, Zuo M, Zhan F, Fan P, Liu S, Taylor M, Ganau M, Hall WA, Ruan H, Wan L. Cognition mediates the relationship between white matter hyperintensity and motor function in patients with cerebral small vessel disease: a cross-sectional study. Quant Imaging Med Surg 2024; 14:7306-7317. [PMID: 39429558 PMCID: PMC11485344 DOI: 10.21037/qims-24-1058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/29/2024] [Indexed: 10/22/2024]
Abstract
Background White matter hyperintensity (WMH) is a common neuroimaging marker of cerebral small vessel disease (SVD) and a critical independent predictor of motor dysfunction, which increases the risk of disability, morbidity, and mortality. However, the mechanism underlying the relationship between WMH and motor function has not yet been fully clarified. It was hypothesized that cognitive impairment mediates the relationship between WMH and motor dysfunction in patients with SVD, which were considered predictor and outcome variables, respectively. Methods A total of 221 patients with SVD were enrolled in this study, and their magnetic resonance imaging (MRI), neuropsychological, and motor function data were collected. The MRI data were visually assessed to determine the WMH burden using the Fazekas scale. Cognition was evaluated using the Montreal Cognitive Assessment (MoCA). Motor function was assessed using the Tinetti Gait and Balance Scale and the Short Physical Performance Battery (SPPB). Finally, a bootstrap analysis was performed to determine whether cognition mediated the relationship between WMH and motor function. Results Of all the patients, 30.3% had mild WMH, 37.6% had moderate WMH, and 32.1% had severe WMH. Patients' cognition and motor function decreased as the WMH burden increased (P<0.01). The MoCA scores were associated with the Tinetti scale (r=0.545, P<0.01) and SPPB scores (r=0.365, P<0.01). Finally, multi-categorical mediation models confirmed our research hypothesis; the coefficients for the indirect effects had 95% confidence intervals (CIs) that excluded zero, indicating statistically significant mediation effects. Conclusions WMH is associated with motor dysfunction, and this association is mediated by cognition in patients with SVD. This finding highlights the importance of early interventions targeting cognitive function to reduce the risk of motor dysfunction.
Collapse
Affiliation(s)
- Xueyang Zhao
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Mengyun Zuo
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fufang Zhan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ping Fan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sanxin Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Marcus Taylor
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Mario Ganau
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Walter A. Hall
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Hengfang Ruan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lihong Wan
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Yohe EO, Alonso A, Drane DL, Patel SS, Schwinne M, Epenge E, Gikelekele G, Herve E, Kavugho I, Tshengele N, Mampunza S, Mananga L, Zhao L, Qiu D, Stringer A, Saindane AM, Ikanga J. Predictors of white matter hyperintensities in the elderly Congolese population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.03.24313022. [PMID: 39281731 PMCID: PMC11398449 DOI: 10.1101/2024.09.03.24313022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background White matter hyperintensities (WMHs) are strongly linked to cardiovascular risk factors and other health conditions such as Alzheimer's disease. However, there is a dearth of research on this topic in low-income countries and underserved populations, especially in the Democratic Republic of Congo (DRC) where the population is aging rapidly with increasing cardiovascular risk factors and dementia-related diseases. This study evaluates health factors associated with WMH in the elderly Sub-Saharan Africa (SSA), specifically Congolese adults. Methods In a cross-sectional study of 77 people from the DRC, participants underwent neuroimaging to analyze WMH volume and completed clinical evaluation, laboratory-based blood exams, self-reported questionnaires, and interviews. A simple linear regression model was conducted to test the association between WMH and potential predictors (neurological status, age, sex, hypertension, diabetes, tobacco abuse, stroke, high cholesterol, cardiovascular medication, and alcohol abuse). Stepwise selection and backward elimination analyses were performed to obtain the final model. Finally, a multiple linear regression model was conducted to assess the association between WMH and variables retained in the final model (neurological status, sex, and age). Results Of the 77 individuals, 47 (61%) had dementia, 40 (52.6%) were males, and the mean age was 73 years (± 8.0 years standard deviation). In simple linear regression models, WMH was significantly associated with dementia (expβ1=1.75, 95% CI=1.14 - 2.71, p-value=0.01) though it had a weak association with age (expβ1=1.03, 95% CI=1.00 - 1.05, p-value=0.05) and sex (male) (expβ1=0.66, 95% CI=0.43 - 1.01, p-value=0.05). In multiple linear regression models, WMH was statistically significantly associated with dementia (expβ1=1.97, 95% CI=1.31 - 2.95, p-value =0.001), male sex (expβ2=0.54, 95% CI=0.36 - 0.80, p-value=0.003), and age (expβ3=1.03, 95% CI=1.00 - 1.06, p-value=0.03). However, WMH was not significantly associated with common cardiovascular risk factors, such as high blood pressure, diabetes, tobacco use, obesity, and high cholesterol levels. Conclusion WMH is significantly associated with neurological status, sex, and age in the Congolese population. Understanding these predictors may improve our ability to diagnose, assess, and develop preventative treatments for white matter disease in SSA/DRC populations, where neuroimaging is difficult to obtain.
Collapse
Affiliation(s)
- Emile Omba Yohe
- Emory University, Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, 30322, USA
| | - Alvaro Alonso
- Emory University, Department of Epidemiology, Rollins School of Public Health, Atlanta, GA, 30322, USA
| | - Daniel L. Drane
- Emory University School of Medicine, Departments of Neurology and Pediatrics, Atlanta, Georgia 30322, USA
| | - Saranya Sundaram Patel
- Emory University School of Medicine, Department of Rehabilitation Medicine, Atlanta, Georgia, 30322, USA
- OneRehab, Dallas, Texas, USA
| | - Megan Schwinne
- Emory University, Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA, USA
| | - Emmanuel Epenge
- University of Kinshasa, Department of Neurology, Kinshasa, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Guy Gikelekele
- University of Kinshasa and Catholic University of Congo, School of Medicine, Kinshasa, Department of Psychiatry, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Esambo Herve
- University of Kinshasa, Department of Neurology, Kinshasa, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Immaculee Kavugho
- Memory clinic of Kinshasa, Kinshasa, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Nathan Tshengele
- University of Kinshasa and Catholic University of Congo, School of Medicine, Kinshasa, Department of Psychiatry, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Samuel Mampunza
- University of Kinshasa and Catholic University of Congo, School of Medicine, Kinshasa, Department of Psychiatry, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Lelo Mananga
- University of Kinshasa, Department of Neurology, Kinshasa, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| | - Liping Zhao
- Emory University, Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA, USA
| | - Deqiang Qiu
- Emory University, School of Medicine, Department of Radiology and Imaging Sciences & Department of Biomedical Engineering, Atlanta, GA, USA
| | - Anthony Stringer
- Emory University School of Medicine, Department of Rehabilitation Medicine, Atlanta, Georgia, 30322, USA
| | - Amit M Saindane
- Emory University School of Medicine, Departments of Radiology and Imaging Sciences and Neurosurgery, Atlanta, GA, USA
| | - Jean Ikanga
- Emory University School of Medicine, Department of Rehabilitation Medicine, Atlanta, Georgia, 30322, USA
- University of Kinshasa and Catholic University of Congo, School of Medicine, Kinshasa, Department of Psychiatry, B.P. 7463 Kinshasa I, Democratic Republic of Congo
- University of Kinshasa and Catholic University of Congo, School of Medicine, Kinshasa, Department of Psychiatry, B.P. 7463 Kinshasa I, Democratic Republic of Congo
| |
Collapse
|
8
|
Solé-Guardia G, Luijten M, Janssen E, Visch R, Geenen B, Küsters B, Claassen JAHR, Litjens G, de Leeuw FE, Wiesmann M, Kiliaan AJ. Deep learning-based segmentation in MRI-(immuno)histological examination of myelin and axonal damage in normal-appearing white matter and white matter hyperintensities. Brain Pathol 2024:e13301. [PMID: 39175459 DOI: 10.1111/bpa.13301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
The major vascular cause of dementia is cerebral small vessel disease (SVD). Its diagnosis relies on imaging hallmarks, such as white matter hyperintensities (WMH). WMH present a heterogenous pathology, including myelin and axonal loss. Yet, these might be only the "tip of the iceberg." Imaging modalities imply that microstructural alterations underlie still normal-appearing white matter (NAWM), preceding the conversion to WMH. Unfortunately, direct pathological characterization of these microstructural alterations affecting myelinated axonal fibers in WMH, and especially NAWM, is still missing. Given that there are no treatments to significantly reduce WMH progression, it is important to extend our knowledge on pathological processes that might already be occurring within NAWM. Staining of myelin with Luxol Fast Blue, while valuable, fails to assess subtle alterations in white matter microstructure. Therefore, we aimed to quantify myelin surrounding axonal fibers and axonal- and microstructural damage in detail by combining (immuno)histochemistry with polarized light imaging (PLI). To study the extent (of early) microstructural damage from periventricular NAWM to the center of WMH, we refined current analysis techniques by using deep learning to define smaller segments of white matter, capturing increasing fluid-attenuated inversion recovery signal. Integration of (immuno)histochemistry and PLI with post-mortem imaging of the brains of individuals with hypertension and normotensive controls enables voxel-wise assessment of the pathology throughout periventricular WMH and NAWM. Myelin loss, axonal integrity, and white matter microstructural damage are not limited to WMH but already occur within NAWM. Notably, we found that axonal damage is higher in individuals with hypertension, particularly in NAWM. These findings highlight the added value of advanced segmentation techniques to visualize subtle changes occurring already in NAWM preceding WMH. By using quantitative MRI and advanced diffusion MRI, future studies may elucidate these very early mechanisms leading to neurodegeneration, which ultimately contribute to the conversion of NAWM to WMH.
Collapse
Affiliation(s)
- Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Matthijs Luijten
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Esther Janssen
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Ruben Visch
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Benno Küsters
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatrics, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Geert Litjens
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
- Computational Pathology Group, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Research Institute for Medical Innovation, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Debette S, Caro I, Western D, Namba S, Sun N, Kawaguchi S, He Y, Fujita M, Roshchupkin G, D'Aoust T, Duperron MG, Sargurupremraj M, Tsuchida A, Koido M, Ahmadi M, Yang C, Timsina J, Ibanez L, Matsuda K, Suzuki Y, Oda Y, Kanai A, Jandaghi P, Munter HM, Auld D, Astafeva I, Puerta R, Rotter J, Psaty B, Bis J, Longstreth W, Couffinhal T, Garcia-Gonzalez P, Pytel V, Marquié M, Cano A, Boada M, Joliot M, Lathrop M, Le Grand Q, Launer L, Wardlaw J, Heiman M, Ruiz A, Matthews P, Seshadri S, Fornage M, Adams H, Mishra A, Trégouët DA, Okada Y, Kellis M, De Jager P, Tzourio C, Kamatani Y, Matsuda F, Cruchaga C. Proteogenomics in cerebrospinal fluid and plasma reveals new biological fingerprint of cerebral small vessel disease. RESEARCH SQUARE 2024:rs.3.rs-4535534. [PMID: 39011113 PMCID: PMC11247936 DOI: 10.21203/rs.3.rs-4535534/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Cerebral small vessel disease (cSVD) is a leading cause of stroke and dementia with no specific mechanism-based treatment. We used Mendelian randomization to combine a unique cerebrospinal fluid (CSF) and plasma pQTL resource with the latest European-ancestry GWAS of MRI-markers of cSVD (white matter hyperintensities, perivascular spaces). We describe a new biological fingerprint of 49 protein-cSVD associations, predominantly in the CSF. We implemented a multipronged follow-up, across fluids, platforms, and ancestries (Europeans and East-Asian), including testing associations of direct plasma protein measurements with MRI-cSVD. We highlight 16 proteins robustly associated in both CSF and plasma, with 24/4 proteins identified in CSF/plasma only. cSVD-proteins were enriched in extracellular matrix and immune response pathways, and in genes enriched in microglia and specific microglial states (integration with single-nucleus RNA sequencing). Immune-related proteins were associated with MRI-cSVD already at age twenty. Half of cSVD-proteins were associated with stroke, dementia, or both, and seven cSVD-proteins are targets for known drugs (used for other indications in directions compatible with beneficial therapeutic effects. This first cSVD proteogenomic signature opens new avenues for biomarker and therapeutic developments.
Collapse
Affiliation(s)
| | | | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory; Broad Institute of MIT and Harvard
| | | | - Yunye He
- Graduate School of Frontier Sciences, The University of Tokyo
| | | | | | - Tim D'Aoust
- Bordeaux Population Health, Inserm U1219, University of Bordeaux
| | | | - Murali Sargurupremraj
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases Unive
| | | | - Masaru Koido
- Graduate School of Frontier Sciences, The University of Tokyo
| | | | | | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo
| | | | - Yoshiya Oda
- Graduate School of Medicine, The University of Tokyo
| | | | | | | | - Dan Auld
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University
| | - Iana Astafeva
- Bordeaux Population Health, Inserm U1219, University of Bordeaux; Institute of Neurodegenerative Diseases
| | | | - Jerome Rotter
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center
| | | | | | | | - Thierry Couffinhal
- University of Bordeaux, The clinical unit of Exploration, Prevention and Care Center for Atherosclerosis (CEPTA), CHUB, Inserm U1034
| | | | - Vanesa Pytel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya; CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III
| | | | | | | | | | - Mark Lathrop
- Department of Human Genetics, McGill University, 1205 Dr Penfield Avenue, Montreal, QC, H3A 1B1, Canada
| | - Quentin Le Grand
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219
| | - Lenore Launer
- National Institute on Aging, National Institutes of Health
| | | | | | - Agustin Ruiz
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center; Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya;CIBERN
| | - Paul Matthews
- UK Dementia Research Institute Centre at Imperial College London
| | | | - Myriam Fornage
- 1. Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center 2. Human Genetics Center, Department of Epidemiology, School of Public Health
| | - Hieab Adams
- Department of Human Genetics, Radboud University Medical Center; Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez
| | | | | | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, The Univ. of Tokyo; Department of Statistical Genetics, Osaka Univ. Graduate School of Medicine; Laboratory for Systems Genetic, RIKEN
| | | | | | | | | | | | | |
Collapse
|
10
|
Solé-Guardia G, Luijten M, Geenen B, Claassen JAHR, Litjens G, de Leeuw FE, Wiesmann M, Kiliaan AJ. Three-dimensional identification of microvascular pathology and neurovascular inflammation in severe white matter hyperintensity: a case report. Sci Rep 2024; 14:5004. [PMID: 38424226 PMCID: PMC10904845 DOI: 10.1038/s41598-024-55733-y] [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: 09/18/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
White matter hyperintensities (WMH) are the most prevalent markers of cerebral small vessel disease (SVD), which is the major vascular risk factor for dementia. Microvascular pathology and neuroinflammation are suggested to drive the transition from normal-appearing white matter (NAWM) to WMH, particularly in individuals with hypertension. However, current imaging techniques cannot capture ongoing NAWM changes. The transition from NAWM into WMH is a continuous process, yet white matter lesions are often examined dichotomously, which may explain their underlying heterogeneity. Therefore, we examined microvascular and neurovascular inflammation pathology in NAWM and severe WMH three-dimensionally, along with gradual magnetic resonance imaging (MRI) fluid-attenuated inversion recovery (FLAIR) signal (sub-)segmentation. In WMH, the vascular network exhibited reduced length and complexity compared to NAWM. Neuroinflammation was more severe in WMH. Vascular inflammation was more pronounced in NAWM, suggesting its potential significance in converting NAWM into WMH. Moreover, the (sub-)segmentation of FLAIR signal displayed varying degrees of vascular pathology, particularly within WMH regions. These findings highlight the intricate interplay between microvascular pathology and neuroinflammation in the transition from NAWM to WMH. Further examination of neurovascular inflammation across MRI-visible alterations could aid deepening our understanding on WMH conversion, and therewith how to improve the prognosis of SVD.
Collapse
Affiliation(s)
- Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Matthijs Luijten
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatrics, Donders Institute for Brain, Cognition & Behavior, Radboud Alzheimer Center, Radboud university medical center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Geert Litjens
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
- Computational Pathology Group, Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition & Behavior, Radboud university medical center, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands.
| |
Collapse
|
11
|
Sheikh AM, Yano S, Tabassum S, Nagai A. The Role of the Vascular System in Degenerative Diseases: Mechanisms and Implications. Int J Mol Sci 2024; 25:2169. [PMID: 38396849 PMCID: PMC10889477 DOI: 10.3390/ijms25042169] [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: 01/12/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Degenerative diseases, encompassing a wide range of conditions affecting various organ systems, pose significant challenges to global healthcare systems. This comprehensive review explores the intricate interplay between the vascular system and degenerative diseases, shedding light on the underlying mechanisms and profound implications for disease progression and management. The pivotal role of the vascular system in maintaining tissue homeostasis is highlighted, as it serves as the conduit for oxygen, nutrients, and immune cells to vital organs and tissues. Due to the vital role of the vascular system in maintaining homeostasis, its dysfunction, characterized by impaired blood flow, endothelial dysfunction, and vascular inflammation, emerges as a common denominator of degenerative diseases across multiple systems. In the nervous system, we explored the influence of vascular factors on neurodegenerative diseases such as Alzheimer's and Parkinson's, emphasizing the critical role of cerebral blood flow regulation and the blood-brain barrier. Within the kidney system, the intricate relationship between vascular health and chronic kidney disease is scrutinized, unraveling the mechanisms by which hypertension and other vascular factors contribute to renal dysfunction. Throughout this review, we emphasize the clinical significance of understanding vascular involvement in degenerative diseases and potential therapeutic interventions targeting vascular health, highlighting emerging treatments and prevention strategies. In conclusion, a profound appreciation of the role of the vascular system in degenerative diseases is essential for advancing our understanding of degenerative disease pathogenesis and developing innovative approaches for prevention and treatment. This review provides a comprehensive foundation for researchers, clinicians, and policymakers seeking to address the intricate relationship between vascular health and degenerative diseases in pursuit of improved patient outcomes and enhanced public health.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
| | - Atsushi Nagai
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.Y.); (S.T.); (A.N.)
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan
| |
Collapse
|
12
|
Weaver DF. Thirty Risk Factors for Alzheimer's Disease Unified by a Common Neuroimmune-Neuroinflammation Mechanism. Brain Sci 2023; 14:41. [PMID: 38248256 PMCID: PMC10813027 DOI: 10.3390/brainsci14010041] [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: 11/29/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
One of the major obstacles confronting the formulation of a mechanistic understanding for Alzheimer's disease (AD) is its immense complexity-a complexity that traverses the full structural and phenomenological spectrum, including molecular, macromolecular, cellular, neurological and behavioural processes. This complexity is reflected by the equally complex diversity of risk factors associated with AD. However, more than merely mirroring disease complexity, risk factors also provide fundamental insights into the aetiology and pathogenesis of AD as a neurodegenerative disorder since they are central to disease initiation and subsequent propagation. Based on a systematic literature assessment, this review identified 30 risk factors for AD and then extended the analysis to further identify neuroinflammation as a unifying mechanism present in all 30 risk factors. Although other mechanisms (e.g., vasculopathy, proteopathy) were present in multiple risk factors, dysfunction of the neuroimmune-neuroinflammation axis was uniquely central to all 30 identified risk factors. Though the nature of the neuroinflammatory involvement varied, the activation of microglia and the release of pro-inflammatory cytokines were a common pathway shared by all risk factors. This observation provides further evidence for the importance of immunopathic mechanisms in the aetiopathogenesis of AD.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Research Institute, University Health Network, Departments of Medicine, Chemistry, Pharmaceutical Sciences, University of Toronto, Toronto, ON M5T 0S8, Canada
| |
Collapse
|
13
|
Morton L, Arndt P, Garza AP, Henneicke S, Mattern H, Gonzalez M, Dityatev A, Yilmazer-Hanke D, Schreiber S, Dunay IR. Spatio-temporal dynamics of microglia phenotype in human and murine cSVD: impact of acute and chronic hypertensive states. Acta Neuropathol Commun 2023; 11:204. [PMID: 38115109 PMCID: PMC10729582 DOI: 10.1186/s40478-023-01672-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/19/2023] [Indexed: 12/21/2023] Open
Abstract
Vascular risk factors such as chronic hypertension are well-established major modifiable factors for the development of cerebral small vessel disease (cSVD). In the present study, our focus was the investigation of cSVD-related phenotypic changes in microglia in human disease and in the spontaneously hypertensive stroke-prone rat (SHRSP) model of cSVD. Our examination of cortical microglia in human post-mortem cSVD cortical tissue revealed distinct morphological microglial features specific to cSVD. We identified enlarged somata, an increase in the territory occupied by thickened microglial processes, and an expansion in the number of vascular-associated microglia. In parallel, we characterized microglia in a rodent model of hypertensive cSVD along different durations of arterial hypertension, i.e., early chronic and late chronic hypertension. Microglial somata were already enlarged in early hypertension. In contrast, at late-stage chronic hypertension, they further exhibited elongated branches, thickened processes, and a reduced ramification index, mirroring the findings in human cSVD. An unbiased multidimensional flow cytometric analysis revealed phenotypic heterogeneity among microglia cells within the hippocampus and cortex. At early-stage hypertension, hippocampal microglia exhibited upregulated CD11b/c, P2Y12R, CD200R, and CD86 surface expression. Detailed analysis of cell subpopulations revealed a unique microglial subset expressing CD11b/c, CD163, and CD86 exclusively in early hypertension. Notably, even at early-stage hypertension, microglia displayed a higher association with cerebral blood vessels. We identified several profound clusters of microglia expressing distinct marker profiles at late chronic hypertensive states. In summary, our findings demonstrate a higher vulnerability of the hippocampus, stage-specific microglial signatures based on morphological features, and cell surface protein expression in response to chronic arterial hypertension. These results indicate the diversity within microglia sub-populations and implicate the subtle involvement of microglia in cSVD pathogenesis.
Collapse
Affiliation(s)
- Lorena Morton
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Philipp Arndt
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Solveig Henneicke
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Faculty of Natural Sciences, Biomedical Magnetic Resonance, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Marilyn Gonzalez
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Deniz Yilmazer-Hanke
- Clinical Neuroanatomy, Department of Neurology, Institute for Biomedical Research, Ulm University, Ulm, Germany
| | - Stefanie Schreiber
- Department of Neurology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Health Campus Immunology, Infectiology, and Inflammation (GC-I3), Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany.
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany.
| |
Collapse
|
14
|
Liu S, Wang M, Gu D, Li Y, Zhang X, Li H, Ji C, Nie X, Liu J. Optimal systolic and diastolic blood pressure threshold that associated with lower risk of white matter hyperintensity progression. Front Aging Neurosci 2023; 15:1254463. [PMID: 37927340 PMCID: PMC10620971 DOI: 10.3389/fnagi.2023.1254463] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Background The optimal control thresholds for systolic blood pressure (SBP) and diastolic blood pressure (DBP) in patients with white matter hyperintensity (WMH) are still unclear. Method A longitudinal retrospective study of patients with brain magnetic resonance imaging (MRI) scans with intervals of more than 3 years was conducted. Blood pressure records during hospitalization and from outpatient visits between baseline and the last MRI scan were collected. The outcome was the change in total WMH from baseline to the final visit. Results Among the 965 patients with MRI scans, 457 patients with detailed longitudinal blood pressure records were ultimately included and classified into the WMH absent group (n = 121), mild WMH group (n = 126), and moderate to severe WMH group (n = 210). Both baseline and longitudinal mean SBP, DBP, and SBP SD were significantly associated with WMH severity (p < 0.05). An average SBP of 130-140 mmHg [vs. <130 mmHg, aOR, 1.80, (95% CI, 1.05-3.07), p = 0.03] was associated with a higher risk of WMH progression. DBP ≥ 90 mmHg [vs. <80 mmHg, OR, 1.81, (95% CI, 0.88-3.74), p = 0.02, aOR, 1.54, (95% CI, 0.66-3.53), p = 0.32] was associated with a higher risk of WMH progression, but was not after adjusted for other covariates. Longitudinal BP variability was not significantly associated with WMH progression. Conclusion Both SBP and DBP had a stronger relationship with the severity of WMH. A target mean SBP of <130 mmHg and mean DBP of <80 mmHg was associated with a lower risk of WMH progression.
Collapse
Affiliation(s)
- Sibo Liu
- Intensive Care Unit, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Mengxing Wang
- China National Clinical Research Center for Neurological Diseases Beijing China, Beijing, China
| | - De’an Gu
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, China
| | - Yanzhao Li
- Department of Neurosurgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xin Zhang
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Hang Li
- Department of Geriatrics, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China
| | - Chenhua Ji
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Ximing Nie
- Neurocritical Care Unit, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinjie Liu
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| |
Collapse
|
15
|
Li L, He G, Shi M, Zhu J, Cheng Y, Chen Y, Chen J, Xue Q. Edaravone dexborneol ameliorates cognitive impairment by regulating the NF-κB pathway through AHR and promoting microglial polarization towards the M2 phenotype in mice with bilateral carotid artery stenosis (BCAS). Eur J Pharmacol 2023; 957:176036. [PMID: 37673366 DOI: 10.1016/j.ejphar.2023.176036] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Cerebral small vessel disease (CSVD) is one of the most important causes of stroke and vascular dementia, so exploring effective treatment modalities for CSVD is warranted. This study aimed to explore the anti-inflammatory effects of Edaravone dexborneol (C.EDA) in a CSVD model. Mice with CSVD showed distinct cognitive decline, as assessed by the Morris water maze (MWM). Pathological staining verified leakage across the blood‒brain barrier (BBB), microglial proliferation, neuronal loss and demyelination. Western blot analysis demonstrated that M1 microglia dominated prophase and released proinflammatory molecules; the aryl hydrocarbon receptor (AHR) was found to participate in modulating nuclear factor-kappa B (NF-κB) signalling activation through tumour necrosis factor receptor-associated factor-6 (TRAF6). C.EDA treatment resulted in the polarization of microglia from the M1 to the M2 phenotype. Mice sequentially treated with C.EDA exhibited a significant improvement in cognitive function; expression of the anti-inflammatory cytokines and modulatory proteins AHR and TRAF6 was upregulated, while the levels of pNF-κBp65 and pIΚBα were downregulated. C.EDA promoted microglial activation towards the M2 phenotype by upregulating AHR expression, which prevented TRAF6 ubiquitination, promoted NF-κB RelA/p65 protein degradation and inhibited subsequent NF-κB phosphorylation. Mechanistically, the anti-inflammatory effect of C.EDA alleviated neuronal loss and myelin damage, while at the functional level, C.EDA improved cognitive function and thus showed good application prospects.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China; Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Guojun He
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Mingyu Shi
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China
| | - Yongqing Cheng
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Yang Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Jin Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
16
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
17
|
Lohkamp KJ, van den Hoek AM, Solé-Guardia G, Lisovets M, Alves Hoffmann T, Velanaki K, Geenen B, Verweij V, Morrison MC, Kleemann R, Wiesmann M, Kiliaan AJ. The Preventive Effect of Exercise and Oral Branched-Chain Amino Acid Supplementation on Obesity-Induced Brain Changes in Ldlr−/−.Leiden Mice. Nutrients 2023; 15:nu15071716. [PMID: 37049556 PMCID: PMC10097391 DOI: 10.3390/nu15071716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Exercise and dietary interventions are promising approaches to tackle obesity and its obesogenic effects on the brain. We investigated the impact of exercise and possible synergistic effects of exercise and branched-chain amino acids (BCAA) supplementation on the brain and behavior in high-fat-diet (HFD)-induced obese Ldlr−/−.Leiden mice. Baseline measurements were performed in chow-fed Ldlr−/−.Leiden mice to assess metabolic risk factors, cognition, and brain structure using magnetic resonance imaging. Thereafter, a subgroup was sacrificed, serving as a healthy reference. The remaining mice were fed an HFD and divided into three groups: (i) no exercise, (ii) exercise, or (iii) exercise and dietary BCAA. Mice were followed for 6 months and aforementioned tests were repeated. We found that exercise alone changed cerebral blood flow, attenuated white matter loss, and reduced neuroinflammation compared to non-exercising HFD-fed mice. Contrarily, no favorable effects of exercise on the brain were found in combination with BCAA, and neuroinflammation was increased. However, cognition was slightly improved in exercising mice on BCAA. Moreover, BCAA and exercise increased the percentage of epididymal white adipose tissue and muscle weight, decreased body weight and fasting insulin levels, improved the circadian rhythm, and transiently improved grip strength. In conclusion, BCAA should be supplemented with caution, although beneficial effects on metabolism, behavior, and cognition were observed.
Collapse
Affiliation(s)
- Klara J. Lohkamp
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Anita M. van den Hoek
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Maria Lisovets
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Talissa Alves Hoffmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Konstantina Velanaki
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (A.M.v.d.H.); (M.C.M.); (R.K.)
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, 6525 EZ Nijmegen, The Netherlands; (K.J.L.); (G.S.-G.); (M.L.); (T.A.H.); (K.V.); (B.G.); (V.V.); (M.W.)
- Correspondence:
| |
Collapse
|