1
|
Li N, Hu YD, Jiang Y, Ling L, Wang CH, Shao JM, Li SB, Di WY. Integrating clinical and biochemical markers: a novel nomogram for predicting lacunes in cerebral small vessel disease. Front Aging Neurosci 2024; 16:1404836. [PMID: 39246593 PMCID: PMC11377284 DOI: 10.3389/fnagi.2024.1404836] [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: 03/21/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Background Lacunes, a characteristic feature of cerebral small vessel disease (CSVD), are critical public health concerns, especially in the aging population. Traditional neuroimaging techniques often fall short in early lacune detection, prompting the need for more precise predictive models. Methods In this retrospective study, 587 patients from the Neurology Department of the Affiliated Hospital of Hebei University who underwent cranial MRI were assessed. A nomogram for predicting lacune incidence was developed using LASSO regression and binary logistic regression analysis for variable selection. The nomogram's performance was quantitatively assessed using AUC-ROC, calibration plots, and decision curve analysis (DCA) in both training (n = 412) and testing (n = 175) cohorts. Results Independent predictors identified included age, gender, history of stroke, carotid atherosclerosis, hypertension, creatinine, and homocysteine levels. The nomogram showed an AUC-ROC of 0.814 (95% CI: 0.791-0.870) for the training set and 0.805 (95% CI: 0.782-0.843) for the testing set. Calibration and DCA corroborated the model's clinical value. Conclusion This study introduces a clinically useful nomogram, derived from binary logistic regression, that significantly enhances the prediction of lacunes in patients undergoing brain MRI for various indications, potentially advancing early diagnosis and intervention. While promising, its retrospective design and single-center context are limitations that warrant further research, including multi-center validation.
Collapse
Affiliation(s)
- Ning Li
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Ya-Dong Hu
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Ye Jiang
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Li Ling
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Chu-Han Wang
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Jia-Min Shao
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Si-Bo Li
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Wei-Ying Di
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| |
Collapse
|
2
|
Wei W, Ma D, Gu L, Li Y, Zhang L, Li L, Zhang L. Epimedium flavonoids improve cerebral white matter lesions by inhibiting neuroinflammation and activating neurotrophic factor signal pathways in spontaneously hypertensive rats. Int Immunopharmacol 2024; 139:112683. [PMID: 39018691 DOI: 10.1016/j.intimp.2024.112683] [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: 05/07/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Cerebral small vessel disease (CSVD) is one of the most common nervous system diseases. Hypertension and neuroinflammation are considered important risk factors for the development of CSVD and white matter (WM) lesions. We used the spontaneously hypertensive rat (SHR) as a model of early-onset CSVD and administered epimedium flavonoids (EF) for three months. The learning and memorization abilities were tested by new object recognition test. The pathological changes of WM were assessed using magnetic resonance imaging, transmission electron microscopy (TEM), Luxol fast blue and Black Gold staining. Oligodendrocytes (OLs) and myelin basic protein were detected by immunohistochemistry. The ultrastructure of the tight junctions was examined using TEM. Microglia and astrocytes were detected by immunofluorescence. RNA-seq was performed on the corpus callosum of rats. The results revealed that EF could significantly improve the learning and memory impairments in SHR, alleviate the injury and demyelination of WM nerve fibers, promote the differentiation of oligodendrocyte precursor cells (OPCs) into mature OLs, inhibit the activation of microglia and astrocytes, inhibit the expression of p38 MAPK/NF-κB p65/NLRP3 and inflammatory cytokines, and increase the expression of tight-junction related proteins ZO-1, occludin, and claudin-5. RNA-seq analysis showed that the neurotrophin signaling pathway played an important role in the disease. RT-qPCR and WB results showed that EF could regulate the expression of nerve growth factor and brain-derived neurotrophic factor and their downstream related proteins in the neurotrophin signaling pathway, which might explain the potential mechanism of EF's effects on the cognitive impairment and WM damage caused by hypertension.
Collapse
Affiliation(s)
- Weipeng Wei
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Denglei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China.
| | - Lihong Gu
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Department of Pharmacy, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yali Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing Engineering Research Center for Nervous System Drugs, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China.
| |
Collapse
|
3
|
Ferris HR, Stine NC, Hill-Eubanks DC, Nelson MT, Wellman GC, Koide M. Epidermal Growth Factor Receptors in Vascular Endothelial Cells Contribute to Functional Hyperemia in the Brain. Int J Mol Sci 2023; 24:16284. [PMID: 38003472 PMCID: PMC10671586 DOI: 10.3390/ijms242216284] [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: 09/15/2023] [Revised: 11/06/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Functional hyperemia-activity-dependent increases in local blood perfusion-underlies the on-demand delivery of blood to regions of enhanced neuronal activity, a process that is crucial for brain health. Importantly, functional hyperemia deficits have been linked to multiple dementia risk factors, including aging, chronic hypertension, and cerebral small vessel disease (cSVD). We previously reported crippled functional hyperemia in a mouse model of genetic cSVD that was likely caused by depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) in capillary endothelial cells (EC) downstream of impaired epidermal growth factor receptor (EGFR) signaling. Here, using EC-specific EGFR-knockout (KO) mice, we directly examined the role of endothelial EGFR signaling in functional hyperemia, assessed by measuring increases in cerebral blood flow in response to contralateral whisker stimulation using laser Doppler flowmetry. Molecular characterizations showed that EGFR expression was dramatically decreased in freshly isolated capillaries from EC-EGFR-KO mice, as expected. Notably, whisker stimulation-induced functional hyperemia was significantly impaired in these mice, an effect that was rescued by administration of PIP2, but not by the EGFR ligand, HB-EGF. These data suggest that the deletion of the EGFR specifically in ECs attenuates functional hyperemia, likely via depleting PIP2 and subsequently incapacitating Kir2.1 channel functionality in capillary ECs. Thus, our study underscores the role of endothelial EGFR signaling in functional hyperemia of the brain.
Collapse
Affiliation(s)
- Hannah R. Ferris
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Nathan C. Stine
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - David C. Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| | - George C. Wellman
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
4
|
Yang Y, Cai X, Zhou M, Chen Y, Pi J, Zhao M, Shi Y, Wang S, Jing J, Chen W, Meng X, Wang Y, Pan Y, Wang Y. Prevalence and Risk Factors of Cerebral Small Vessel Disease from a Population-Based Cohort in China. Neuroepidemiology 2023; 57:413-422. [PMID: 37734325 DOI: 10.1159/000533678] [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: 02/12/2023] [Accepted: 07/25/2023] [Indexed: 09/23/2023] Open
Abstract
INTRODUCTION Cerebral small vessel disease (CSVD) is a significant burden of morbidity and mortality among elderly people around the world. Epidemiological data with complete CSVD evaluations and a large sample size in the general population are still limited. METHODS Community-dwelling residents in Lishui city in China from the cross-sectional survey of the Polyvascular Evaluation for Cognitive Impairment and Vascular Events (PRECISE) study were included in this study from 2017 to 2019. All participants underwent 3 Tesla brain magnetic resonance images to assess CSVD imaging markers. Demographic and risk factor data were collected. The general and age-specific prevalence of lacune, confluent white matter hyperintensity (WMH), moderate-severe enlarged perivascular spaces (EPVS), cerebral microbleed (CMB), and total CSVD score (an ordinal scale from 0 to 4, counting the presence of four imaging markers of CSVD) was evaluated. Associations between vascular risk factors and these markers were analyzed by multivariable logistic regression. RESULTS A total of 3,063 participants were enrolled. The mean age was 61.2 years and 46.5% were men. The most prevalent CSVD marker was confluent WMH (16.7%), followed by CMB (10.2%), moderate-severe EPVS in the basal ganglia (BG-EPVS) (9.8%), and lacune (5.6%). 30.5% of the participants have at least one of the four markers (total CSVD score ≥1 points). The prevalence of CSVD markers increases as age increases. Age and hypertension were independent risk factors for four CSVD markers and the total CSVD score. CONCLUSIONS In this Chinese cohort with community-based adults aged 50-75 years, our findings showed a prevalence of 30.5% for CSVD. The most prevalent CSVD marker was confluent WMH, followed by CMB, moderate-severe BG-EPVS, and lacune. The risk factors for CSVD must be strictly screened and controlled in adults living in the community.
Collapse
Affiliation(s)
- Yingying Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,
- China National Clinical Research Center for Neurological Diseases, Beijing, China,
| | - Xueli Cai
- Department of Neurology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Mengyuan Zhou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yiyi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jingtao Pi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mengxi Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yulu Shi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Suying Wang
- Cerebrovascular Research Lab, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Weiqi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
5
|
Negative correlation between serum pyruvate kinase M2 and cognitive function in patients with cerebral small vessel disease. Clin Neurol Neurosurg 2023; 225:107586. [PMID: 36641992 DOI: 10.1016/j.clineuro.2023.107586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Cerebral small vessel disease (CSVD) is one of the main contributing factors to vascular cognitive impairment (VCI), with an increasing incidence rate. However, the genesis of CSVD cognitive impairment remains unknown. Inflammation and metabolic disorders are considered important pathogenesis of CSVD. In addition to acting as the key regulator of aerobic glycolysis, pyruvate kinase muscle isozyme 2 (PKM2) is a proinflammatory mediator transcriptional activator that can promote an inflammatory response. This study explored whether serum PKM2 is associated with cognitive impairment in CSVD patients. METHODS The demographic data, history of risk factors, laboratory data, and cognitive function scale assessment of 219 CSVD patients were analyzed, and the correlation between the CSVD clinical data and neuroimaging parameters with serum PKM2 was further explored. The serum PKM2 level was determined by enzyme-linked immunosorbent assay using the collected serum samples. Insulin resistance (IR) was assessed with reference to the Homeostasis Model Assessment of Insulin Resistance (HOMA-IR). HOMA-IR was calculated using the formula HOMA-IR = fasting plasma glucose (FPG, mmol/L) × fasting insulin (FINS, μU/mL)/22.5. A binomial logistic regression model was referred to infer the risk factors for VCI, and the ability of serum PKM2 to diagnose VCI was assessed by using a ROC curve. RESULTS Serum PKM2 level was positively correlated with HOMA-IR (r = 0.206, P = 0.002), negatively correlated with MMSE and MOCA on the cognitive scale in CSVD patients, and higher in CSVD patients with white matter hyperintensities (WMH) (P < 0.001). When compared with patients without cognitive impairment, the serum PKM2 levels were elevated in cases with suspected dementia, mild dementia, mild to moderate dementia, and moderate to severe dementia, and the differences were statistically significant (P < 0.05). Serum PKM2 levels were correlated with cognitive screening test scores in CSVD. CONCLUSION The present findings indicated that the serum PKM2 level was positively correlated with HOMA-IR, WMH, and enlarged perivascular spaces and negatively correlated with cognitive function in CSVD patients.
Collapse
|
6
|
Oliveira DV, Coupland KG, Shao W, Jin S, Del Gaudio F, Wang S, Fox R, Rutten JW, Sandin J, Zetterberg H, Lundkvist J, Lesnik Oberstein SAJ, Lendahl U, Karlström H. Active immunotherapy reduces NOTCH3 deposition in brain capillaries in a CADASIL mouse model. EMBO Mol Med 2022; 15:e16556. [PMID: 36524456 PMCID: PMC9906330 DOI: 10.15252/emmm.202216556] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/14/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic form of familial small vessel disease; no preventive or curative therapy is available. CADASIL is caused by mutations in the NOTCH3 gene, resulting in a mutated NOTCH3 receptor, with aggregation of the NOTCH3 extracellular domain (ECD) around vascular smooth muscle cells. In this study, we have developed a novel active immunization therapy specifically targeting CADASIL-like aggregated NOTCH3 ECD. Immunizing CADASIL TgN3R182C150 mice with aggregates composed of CADASIL-R133C mutated and wild-type EGF1-5 repeats for a total of 4 months resulted in a marked reduction (38-48%) in NOTCH3 deposition around brain capillaries, increased microglia activation and lowered serum levels of NOTCH3 ECD. Active immunization did not impact body weight, general behavior, the number and integrity of vascular smooth muscle cells in the retina, neuronal survival, or inflammation or the renal system, suggesting that the therapy is tolerable. This is the first therapeutic study reporting a successful reduction of NOTCH3 accumulation in a CADASIL mouse model supporting further development towards clinical application for the benefit of CADASIL patients.
Collapse
Affiliation(s)
- Daniel V Oliveira
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
| | - Kirsten G Coupland
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Wenchao Shao
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Shaobo Jin
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| | | | - Sailan Wang
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Rhys Fox
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| | - Julie W Rutten
- Department of Clinical GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Johan Sandin
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Alzecure FoundationHuddingeSweden,Alzecure PharmaHuddingeSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK,UK Dementia Research Institute at UCLLondonUK,Hong Kong Center for Neurodegenerative Diseases, Clear Water BayHong KongChina
| | - Johan Lundkvist
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Alzecure FoundationHuddingeSweden,Sinfonia BiotherapeuticsHuddingeSweden
| | | | - Urban Lendahl
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden,Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| | - Helena Karlström
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| |
Collapse
|
7
|
Kaya T, Mattugini N, Liu L, Ji H, Cantuti-Castelvetri L, Wu J, Schifferer M, Groh J, Martini R, Besson-Girard S, Kaji S, Liesz A, Gokce O, Simons M. CD8 + T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat Neurosci 2022; 25:1446-1457. [PMID: 36280798 PMCID: PMC9630119 DOI: 10.1038/s41593-022-01183-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 09/14/2022] [Indexed: 01/13/2023]
Abstract
A hallmark of nervous system aging is a decline of white matter volume and function, but the underlying mechanisms leading to white matter pathology are unknown. In the present study, we found age-related alterations of oligodendrocyte cell state with a reduction in total oligodendrocyte density in aging murine white matter. Using single-cell RNA-sequencing, we identified interferon (IFN)-responsive oligodendrocytes, which localize in proximity to CD8+ T cells in aging white matter. Absence of functional lymphocytes decreased the number of IFN-responsive oligodendrocytes and rescued oligodendrocyte loss, whereas T-cell checkpoint inhibition worsened the aging response. In addition, we identified a subpopulation of lymphocyte-dependent, IFN-responsive microglia in the vicinity of the CD8+ T cells in aging white matter. In summary, we provide evidence that CD8+ T-cell-induced, IFN-responsive oligodendrocytes and microglia are important modifiers of white matter aging.
Collapse
Affiliation(s)
- Tuğberk Kaya
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Nicola Mattugini
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Lu Liu
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jianping Wu
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Janos Groh
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf Martini
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Seiji Kaji
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany.
- Munich Cluster of Systems Neurology, Munich, Germany.
| | - Mikael Simons
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany.
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster of Systems Neurology, Munich, Germany.
| |
Collapse
|
8
|
Muratoglu SC, Charette MF, Galis ZS, Greenstein AS, Daugherty A, Joutel A, Kozel BA, Wilcock DM, Collins EC, Sorond FA, Howell GR, Hyacinth HI, Lloyd KKC, Stenmark KR, Boehm M, Kahn ML, Corriveau R, Wells S, Bussey TJ, Sukoff Rizzo SJ, Iruela-Arispe ML. Perspectives on Cognitive Phenotypes and Models of Vascular Disease. Arterioscler Thromb Vasc Biol 2022; 42:831-838. [PMID: 35510549 PMCID: PMC9233038 DOI: 10.1161/atvbaha.122.317395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical investigations have established that vascular-associated medical conditions are significant risk factors for various kinds of dementia. And yet, we are unable to associate certain types of vascular deficiencies with specific cognitive impairments. The reasons for this are many, not the least of which are that most vascular disorders are multi-factorial and the development of vascular dementia in humans is often a multi-year or multi-decade progression. To better study vascular disease and its underlying causes, the National Heart, Lung, and Blood Institute of the National Institutes of Health has invested considerable resources in the development of animal models that recapitulate various aspects of human vascular disease. Many of these models, mainly in the mouse, are based on genetic mutations, frequently using single-gene mutations to examine the role of specific proteins in vascular function. These models could serve as useful tools for understanding the association of specific vascular signaling pathways with specific neurological and cognitive impairments related to dementia. To advance the state of the vascular dementia field and improve the information sharing between the vascular biology and neurobehavioral research communities, National Heart, Lung, and Blood Institute convened a workshop to bring in scientists from these knowledge domains to discuss the potential utility of establishing a comprehensive phenotypic cognitive assessment of a selected set of existing mouse models, representative of the spectrum of vascular disorders, with particular attention focused on age, sex, and rigor and reproducibility. The workshop highlighted the potential of associating well-characterized vascular disease models, with validated cognitive outcomes, that can be used to link specific vascular signaling pathways with specific cognitive and neurobehavioral deficits.
Collapse
Affiliation(s)
- Selen C Muratoglu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (S.C.M., M.F.C., Z.S.G.)
| | - Marc F Charette
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (S.C.M., M.F.C., Z.S.G.)
| | - Zorina S Galis
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (S.C.M., M.F.C., Z.S.G.)
| | - Adam S Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom (A.S.G.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université Paris Descartes, France (A.J.)
| | - Beth A Kozel
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (B.A.K., M.B.)
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience (D.M.W.), University of Kentucky, Lexington
| | | | - Farzaneh A Sorond
- Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL (F.A.S.)
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME (G.R.H.)
- Graduate Program of Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA (G.R.H.)
| | - Hyacinth I Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH (H.I.H.)
| | - Kent K C Lloyd
- Mutant Mouse Resource and Research Center (MMRRC) at the University of California, Davis (K.K.C.L.)
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver (K.R.S.)
| | - Manfred Boehm
- Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (B.A.K., M.B.)
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia (M.L.K.)
| | - Roderick Corriveau
- National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (R.C.)
| | - Sara Wells
- Mary Lyon Centre, Harwell Campus, MRC Harwell Institute, Oxfordshire, United Kingdom (S.W.)
| | - Timothy J Bussey
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (T.J.B.)
| | - Stacey J Sukoff Rizzo
- Department of Medicine-Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA (S.J.S.R.)
| | - M Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL (M.L.I.-A.)
| |
Collapse
|
9
|
A Novel Rodent Model of Hypertensive Cerebral Small Vessel Disease with White Matter Hyperintensities and Peripheral Oxidative Stress. Int J Mol Sci 2022; 23:ijms23115915. [PMID: 35682594 PMCID: PMC9180536 DOI: 10.3390/ijms23115915] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is the second most common cause of stroke and a major contributor to dementia. Manifestations of CSVD include cerebral microbleeds, intracerebral hemorrhages (ICH), lacunar infarcts, white matter hyperintensities (WMH) and enlarged perivascular spaces. Chronic hypertensive models have been found to reproduce most key features of the disease. Nevertheless, no animal models have been identified to reflect all different aspects of the human disease. Here, we described a novel model for CSVD using salt-sensitive ‘Sabra’ hypertension-prone rats (SBH/y), which display chronic hypertension and enhanced peripheral oxidative stress. SBH/y rats were either administered deoxycorticosteroid acetate (DOCA) (referred to as SBH/y-DOCA rats) or sham-operated and provided with 1% NaCl in drinking water. Rats underwent neurological assessment and behavioral testing, followed by ex vivo MRI and biochemical and histological analyses. SBH/y-DOCA rats show a neurological decline and cognitive impairment and present multiple cerebrovascular pathologies associated with CSVD, such as ICH, lacunes, enlarged perivascular spaces, blood vessel stenosis, BBB permeability and inflammation. Remarkably, SBH/y-DOCA rats show severe white matter pathology as well as WMH, which are rarely reported in commonly used models. Our model may serve as a novel platform for further understanding the mechanisms underlying CSVD and for testing novel therapeutics.
Collapse
|
10
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 493] [Impact Index Per Article: 164.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
11
|
Koide M, Harraz OF, Dabertrand F, Longden TA, Ferris HR, Wellman GC, Hill-Eubanks DC, Greenstein AS, Nelson MT. Differential restoration of functional hyperemia by antihypertensive drug classes in hypertension-related cerebral small vessel disease. J Clin Invest 2021; 131:e149029. [PMID: 34351870 PMCID: PMC8439604 DOI: 10.1172/jci149029] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/28/2021] [Indexed: 02/05/2023] Open
Abstract
Dementia resulting from small vessel diseases (SVDs) of the brain is an emerging epidemic for which there is no treatment. Hypertension is the major risk factor for SVDs, but how hypertension damages the brain microcirculation is unclear. Here, we show that chronic hypertension in a mouse model progressively disrupts on-demand delivery of blood to metabolically active areas of the brain (functional hyperemia) through diminished activity of the capillary endothelial cell inward-rectifier potassium channel, Kir2.1. Despite similar efficacy in reducing blood pressure, amlodipine, a voltage-dependent calcium-channel blocker, prevented hypertension-related damage to functional hyperemia whereas losartan, an angiotensin II type 1 receptor blocker, did not. We attribute this drug class effect to losartan-induced aldosterone breakthrough, a phenomenon triggered by pharmacological interruption of the renin-angiotensin pathway leading to elevated plasma aldosterone levels. This hypothesis is supported by the finding that combining losartan with the aldosterone receptor antagonist eplerenone prevented the hypertension-related decline in functional hyperemia. Collectively, these data suggest Kir2.1 as a possible therapeutic target in vascular dementia and indicate that concurrent mineralocorticoid aldosterone receptor blockade may aid in protecting against late-life cognitive decline in hypertensive patients treated with angiotensin II type 1 receptor blockers.
Collapse
Affiliation(s)
- Masayo Koide
- Department of Pharmacology, Larner College of Medicine, and,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA
| | - Osama F. Harraz
- Department of Pharmacology, Larner College of Medicine, and,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA
| | - Fabrice Dabertrand
- Department of Pharmacology, Larner College of Medicine, and,Department of Anesthesiology and,Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas A. Longden
- Department of Pharmacology, Larner College of Medicine, and,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Adam S. Greenstein
- Department of Pharmacology, Larner College of Medicine, and,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA.,Division of Cardiovascular Sciences, School of Medical Sciences and,Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, United Kingdom
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine, and,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA.,Division of Cardiovascular Sciences, School of Medical Sciences and,Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, United Kingdom
| |
Collapse
|