1
|
Kim GH, Kim BR, Yoon HJ, Jeong JH. Elevated cerebral blood flow proxy with increased beta-amyloid burden in Alzheimer's disease preclinical phase evaluated by dual-phase 18F-florbetaben positron emission tomography. Sci Rep 2024; 14:18480. [PMID: 39122860 PMCID: PMC11315901 DOI: 10.1038/s41598-024-68916-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
This study investigated the earliest change of cerebral blood flow (CBF) and its relationship with β-amyloid (Aβ) burden in preclinical Alzheimer's disease (AD) employing dual-phase 18F-florbetaben (FBB) PET. Seventy-one cognitively normal (NC) individuals were classified as Aβ negative (Aβ-NC) or positive (Aβ+NC) based on two different cutoff values: an SUVR of > 1.08 and a Centiloid scale of > 20. The PET scans were acquired in two phases: an early phase (0-10 min, eFBB) and a delayed phase (90-110 min, dFBB), which were averaged to generate single-frame images for each phase. Furthermore, an R1 parametric map was generated from the early phase data using a simplified reference tissue model. We conducted regional and voxel-based analyses to compare the eFBB, dFBB, and R1 images between the Aβ positive and negative groups. In addition, the correlations between the CBF proxy R1 and the dFBB SUVR were analyzed. The Aβ+NC group showed significantly higher dFBB SUVR in both the global cerebral cortex and target regions compared to the Aβ-NC group, while no significant differences were observed in eFBB SUVR between the two groups. Furthermore, the Aβ+NC group exhibited significantly higher R1 values, a proxy for cerebral perfusion, in both the global cerebral cortex and target regions compared to the Aβ-NC group. Significant positive correlations were observed between R1 and dFBB SUVR in both the global cerebral cortex and target regions, which remained significant after controlling for demographics and cognitive profiles, except for the medial temporal and occipital cortices. The findings reveal increased CBF in preclinical AD and a positive correlation between CBF and amyloid pathology. The positive correlation between R1 and amyloid burden may indicate a compensatory mechanism in the preclinical stage of Alzheimer's disease, but to elucidate this hypothesis, further longitudinal observational studies are necessary.
Collapse
Affiliation(s)
- Geon Ha Kim
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Bori R Kim
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Republic of Korea
- Ewha Medical Research Institute, Ewha Womans University, Seoul, Republic of Korea
| | - Hai-Jeon Yoon
- Department of Nuclear Medicine, Ewha Womans University, College of Medicine, Seoul, Republic of Korea.
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Uekawa K, Anfray A, Ahn SJ, Casey N, Seo J, Zhou P, Iadecola C, Park L. tPA supplementation preserves neurovascular and cognitive function in Tg2576 mice. Alzheimers Dement 2024; 20:4572-4582. [PMID: 38899570 PMCID: PMC11247712 DOI: 10.1002/alz.13878] [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: 02/16/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Amyloid beta (Aβ) impairs the cerebral blood flow (CBF) increase induced by neural activity (functional hyperemia). Tissue plasminogen activator (tPA) is required for functional hyperemia, and in mouse models of Aβ accumulation tPA deficiency contributes to neurovascular and cognitive impairment. However, it remains unknown if tPA supplementation can rescue Aβ-induced neurovascular and cognitive dysfunction. METHODS Tg2576 mice and wild-type littermates received intranasal tPA (0.8 mg/kg/day) or vehicle 5 days a week starting at 11 to 12 months of age and were assessed 3 months later. RESULTS Treatment of Tg2576 mice with tPA restored resting CBF, prevented the attenuation in functional hyperemia, and improved nesting behavior. These effects were associated with reduced cerebral atrophy and cerebral amyloid angiopathy, but not parenchymal amyloid. DISCUSSION These findings highlight the key role of tPA deficiency in the neurovascular and cognitive dysfunction associated with amyloid pathology, and suggest potential therapeutic strategies involving tPA reconstitution. HIGHLIGHTS Amyloid beta (Aβ) induces neurovascular dysfunction and impairs the increase of cerebral blood flow induced by neural activity (functional hyperemia). Tissue plasminogen activator (tPA) deficiency contributes to the neurovascular and cognitive dysfunction caused by Aβ. In mice with florid amyloid pathology intranasal administration of tPA rescues the neurovascular and cognitive dysfunction and reduces brain atrophy and cerebral amyloid angiopathy. tPA deficiency plays a crucial role in neurovascular and cognitive dysfunction induced by Aβ and tPA reconstitution may be of therapeutic value.
Collapse
Affiliation(s)
- Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - James Seo
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
3
|
Chen HC, Cao JX, Zhang YS, Ma YZ, Zhang L, Su XM, Gao LP, Jing YH. High salt diet exacerbates cognitive deficits and neurovascular abnormalities in APP/PS1 mice and induces AD-like changes in wild-type mice. J Nutr Biochem 2024; 125:109570. [PMID: 38218348 DOI: 10.1016/j.jnutbio.2024.109570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/25/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
High salt diet (HSD) is a risk factor of hypertension and cardiovascular disease. Although clinical data do not clearly indicate the relationship between HSD and the prevalence of Alzheimer's disease (AD), animal experiments have shown that HSD can cause hyperphosphorylation of tau protein and cognition impairment. However, whether HSD can accelerate the progression of AD by damaging the function of neurovascular unit (NVU) in the brain is unclear. Here, we fed APP/PS1 mice (an AD model) or wild-type mice with HSD and found that the chronic HSD feeding increased the activity of enzymes related to tau phosphorylation, which led to tau hyperphosphorylation in the brain. HSD also aggravated the deposition of Aβ42 in hippocampus and cortex in the APP/PS1 mice but not in the wild-type mice. Simultaneously, HSD caused the microglia proliferation, low expression of Aqp-4, and high expression of CD31 in the wild-type mice, which were accompanied with the loss of pericytes (PCs) and increase in blood brain barrier (BBB) permeability. As a result, wild-type mice fed with HSD performed poorly in Morris Water Maze and object recognition test. In the APP/PS1 mice, HSD feeding for 8 months worsen the cognition and accompanied the loss of PCs, the activation of glia, the increase in BBB permeability, and the acceleration of calcification in the brain. Our data suggested that HSD feeding induced the AD-like pathology in wild-type mice and aggravated the development of AD-like pathology in APP/PS1 mice, which implicated the tau hyperphosphorylation and NVU dysfunction.
Collapse
Affiliation(s)
- Hai Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Lu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
4
|
Fang X, Fan F, Border JJ, Roman RJ. Cerebrovascular Dysfunction in Alzheimer's Disease and Transgenic Rodent Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2024; 5:42-64. [PMID: 38434588 PMCID: PMC10906803 DOI: 10.33696/neurol.5.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Alzheimer's Disease (AD) and Alzheimer's Disease-Related Dementia (ADRD) are the primary causes of dementia that has a devastating effect on the quality of life and is a tremendous economic burden on the healthcare system. The accumulation of extracellular beta-amyloid (Aβ) plaques and intracellular hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain are the hallmarks of AD. They are also thought to be the underlying cause of inflammation, neurodegeneration, brain atrophy, and cognitive impairments that accompany AD. The discovery of APP, PS1, and PS2 mutations that increase Aβ production in families with early onset familial AD led to the development of numerous transgenic rodent models of AD. These models have provided new insight into the role of Aβ in AD; however, they do not fully replicate AD pathology in patients. Familial AD patients with mutations that elevate the production of Aβ represent only a small fraction of dementia patients. In contrast, those with late-onset sporadic AD constitute the majority of cases. This observation, along with the failure of previous clinical trials targeting Aβ or Tau and the modest success of recent trials using Aβ monoclonal antibodies, has led to a reappraisal of the view that Aβ accumulation is the sole factor in the pathogenesis of AD. More recent studies have established that cerebral vascular dysfunction is one of the earliest changes seen in AD, and 67% of the candidate genes linked to AD are expressed in the cerebral vasculature. Thus, there is an increasing appreciation of the vascular contribution to AD, and the National Institute on Aging (NIA) and the Alzheimer's Disease Foundation recently prioritized it as a focused research area. This review summarizes the strengths and limitations of the most commonly used transgenic AD animal models and current views about the contribution of Aβ accumulation versus cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Physiology, Augusta University, Augusta, GA 30912, USA
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
5
|
Floriddia E. In conversation with Costantino Iadecola. Nat Neurosci 2023; 26:2042-2045. [PMID: 37973870 DOI: 10.1038/s41593-023-01505-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
|
6
|
Carballo Á, López-Dequidt I, Custodia A, Botelho J, Aramburu-Núñez M, Machado V, Pías-Peleteiro JM, Ouro A, Romaus-Sanjurjo D, Vázquez-Vázquez L, Jiménez-Martín I, Aguiar P, Rodríguez-Yáñez M, Aldrey JM, Blanco J, Castillo J, Sobrino T, Leira Y. Association of periodontitis with cognitive decline and its progression: Contribution of blood-based biomarkers of Alzheimer's disease to this relationship. J Clin Periodontol 2023; 50:1444-1454. [PMID: 37584311 DOI: 10.1111/jcpe.13861] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023]
Abstract
AIM To assess whether periodontitis is associated with cognitive decline and its progression as well as with certain blood-based markers of Alzheimer's disease. MATERIALS AND METHODS Data from a 2-year follow-up prospective cohort study (n = 101) was analysed. Participants with a previous history of hypertension and aged ≥60 years were included in the analysis. All of them received a full-mouth periodontal examination and cognitive function assessments (Addenbrooke's Cognitive Examination (ACE) and Mini-Mental State Examination [MMSE]). Plasma levels of amyloid beta (Aβ)1-40 , Aβ1-42 , phosphorylated and total Tau (p-Tau and t-Tau) were determined at baseline, 12 and 24 months. RESULTS Periodontitis was associated with poor cognitive performance (MMSE: β = -1.5 [0.6]) and progression of cognitive impairment (hazard ratio [HR] = 1.8; 95% confidence interval: 1.0-3.1). Subjects with periodontitis showed greater baseline levels of p-Tau (1.6 [0.7] vs. 1.2 [0.2] pg/mL, p < .001) and Aβ1-40 (242.1 [77.3] vs. 208.2 [73.8] pg/mL, p = .036) compared with those without periodontitis. Concentrations of the latter protein also increased over time only in the periodontitis group (p = .005). CONCLUSIONS Periodontitis is associated with cognitive decline and its progression in elderly patients with a previous history of hypertension. Overexpression of p-Tau and Aβ1-40 may play a role in this association.
Collapse
Affiliation(s)
- Álvaro Carballo
- Periodontology Unit, Faculty of Odontology and Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Stroke Unit, Neurology Department, University Clinical Hospital, Santiago de Compostela, Spain
| | - Antía Custodia
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - João Botelho
- Periodontology Department and Evidence-Based Hub, Clinical Research Unit, Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz - Cooperativa de Ensino Superior, Caparica, Portugal
| | - Marta Aramburu-Núñez
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - Vanessa Machado
- Periodontology Department and Evidence-Based Hub, Clinical Research Unit, Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz - Cooperativa de Ensino Superior, Caparica, Portugal
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
- Dementia Unit, Neurology Department, University Clinical Hospital, Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - Laura Vázquez-Vázquez
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - Isabel Jiménez-Martín
- Dementia Unit, Neurology Department, University Clinical Hospital, Santiago de Compostela, Spain
| | - Pablo Aguiar
- Molecular Imaging Group, Department of Radiology, Faculty of Medicine and Center for Research In Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Nuclear Medicine Department and Molecular Imaging Group, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Neurology Department, University Clinical Hospital, Santiago de Compostela, Spain
| | - José Manuel Aldrey
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
- Dementia Unit, Neurology Department, University Clinical Hospital, Santiago de Compostela, Spain
| | - Juan Blanco
- Periodontology Unit, Faculty of Odontology and Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL) Group, Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
| | - Tomás Sobrino
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| | - Yago Leira
- Periodontology Unit, Faculty of Odontology and Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital, Santiago de Compostela, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Uekawa K, Hattori Y, Ahn SJ, Seo J, Casey N, Anfray A, Zhou P, Luo W, Anrather J, Park L, Iadecola C. Border-associated macrophages promote cerebral amyloid angiopathy and cognitive impairment through vascular oxidative stress. Mol Neurodegener 2023; 18:73. [PMID: 37789345 PMCID: PMC10548599 DOI: 10.1186/s13024-023-00660-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a devastating condition common in patients with Alzheimer's disease but also observed in the general population. Vascular oxidative stress and neurovascular dysfunction have been implicated in CAA but the cellular source of reactive oxygen species (ROS) and related signaling mechanisms remain unclear. We tested the hypothesis that brain border-associated macrophages (BAM), yolk sac-derived myeloid cells closely apposed to parenchymal and leptomeningeal blood vessels, are the source of radicals through the Aβ-binding innate immunity receptor CD36, leading to neurovascular dysfunction, CAA, and cognitive impairment. METHODS Tg2576 mice and WT littermates were transplanted with CD36-/- or CD36+/+ bone marrow at 12-month of age and tested at 15 months. This approach enables the repopulation of perivascular and leptomeningeal compartments with CD36-/- BAM. Neurovascular function was tested in anesthetized mice equipped with a cranial window in which cerebral blood flow was monitored by laser-Doppler flowmetry. Amyloid pathology and cognitive function were also examined. RESULTS The increase in blood flow evoked by whisker stimulation (functional hyperemia) or by endothelial and smooth muscle vasoactivity was markedly attenuated in WT → Tg2576 chimeras but was fully restored in CD36-/- → Tg2576 chimeras, in which BAM ROS production was suppressed. CAA-associated Aβ1-40, but not Aβ1-42, was reduced in CD36-/- → Tg2576 chimeras. Similarly, CAA, but not parenchymal plaques, was reduced in CD36-/- → Tg2576 chimeras. These beneficial vascular effects were associated with cognitive improvement. Finally, CD36-/- mice were able to more efficiently clear exogenous Aβ1-40 injected into the neocortex or the striatum. CONCLUSIONS CD36 deletion in BAM suppresses ROS production and rescues the neurovascular dysfunction and damage induced by Aβ. CD36 deletion in BAM also reduced brain Aβ1-40 and ameliorated CAA without affecting parenchyma plaques. Lack of CD36 enhanced the vascular clearance of exogenous Aβ. Restoration of neurovascular function and attenuation of CAA resulted in a near complete rescue of cognitive function. Collectively, these data implicate brain BAM in the pathogenesis of CAA and raise the possibility that targeting BAM CD36 is beneficial in CAA and other conditions associated with vascular Aβ deposition and damage.
Collapse
Affiliation(s)
- Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - James Seo
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Wenjie Luo
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
8
|
Fang X, Border JJ, Rivers PL, Zhang H, Williams JM, Fan F, Roman RJ. Amyloid beta accumulation in TgF344-AD rats is associated with reduced cerebral capillary endothelial Kir2.1 expression and neurovascular uncoupling. GeroScience 2023; 45:2909-2926. [PMID: 37326915 PMCID: PMC10643802 DOI: 10.1007/s11357-023-00841-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023] Open
Abstract
Alzheimer's disease (AD) exerts a tremendous socio-economic burden worldwide. Although reduced cerebral blood flow is an early and persistent symptom that precedes the loss of cognitive function in AD, the underlying molecular and cellular mechanisms remain unclear. The present study investigated whether capillary endothelial inward rectifier potassium 2 (Kir2.1) expression is reduced in TgF344-AD (AD) rats and contributes to neurovascular uncoupling and cognitive deficits in AD. Three- to fourteen-month-old AD rats expressing mutant human APP and PS1 and age-matched wild-type (WT) F344 rats were studied. AD rats exhibited higher amyloid beta (Aβ) expression in the brain as early as 3 months of age and amyloid plaques by 4 months of age. Functional hyperemic responses induced by whisker stimulation were impaired at 4 months of age, which were exacerbated in 6-month- and 14-month-old AD rats. The expression of Kir2.1 protein was significantly lower in the brains of 6-month-old AD versus WT rats, and Kir2.1 coverage was lower in the cerebral microvasculature of AD than in WT rats. Aβ1-42 reduced the Kir2.1 expression in cultured capillary endothelial cells. Cerebral parenchymal arterioles with attached capillaries exhibited a reduced vasodilator in response to 10 mM K+ applied to capillaries, and constricted less following administration of a Kir2.1 channel blocker, compared to WT vessels. These results indicate that capillary endothelial Kir2.1 expression is reduced and contributes to impaired functional hyperemia in AD rats at early ages, perhaps secondary to elevated Aβ expression.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Patrice L Rivers
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jan Michael Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
9
|
Ahn SJ, Anfray A, Anrather J, Iadecola C. Calcium transients in nNOS neurons underlie distinct phases of the neurovascular response to barrel cortex activation in awake mice. J Cereb Blood Flow Metab 2023; 43:1633-1647. [PMID: 37149758 PMCID: PMC10581240 DOI: 10.1177/0271678x231173175] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/14/2023] [Accepted: 04/02/2023] [Indexed: 05/08/2023]
Abstract
Neuronal nitric oxide (NO) synthase (nNOS), a Ca2+ dependent enzyme, is expressed by distinct populations of neocortical neurons. Although neuronal NO is well known to contribute to the blood flow increase evoked by neural activity, the relationships between nNOS neurons activity and vascular responses in the awake state remain unclear. We imaged the barrel cortex in awake, head-fixed mice through a chronically implanted cranial window. The Ca2+ indicator GCaMP7f was expressed selectively in nNOS neurons using adenoviral gene transfer in nNOScre mice. Air-puffs directed at the contralateral whiskers or spontaneous motion induced Ca2+ transients in 30.2 ± 2.2% or 51.6 ± 3.3% of nNOS neurons, respectively, and evoked local arteriolar dilation. The greatest dilatation (14.8 ± 1.1%) occurred when whisking and motion occurred simultaneously. Ca2+ transients in individual nNOS neurons and local arteriolar dilation showed various degrees of correlation, which was strongest when the activity of whole nNOS neuron ensemble was examined. We also found that some nNOS neurons became active immediately prior to arteriolar dilation, while others were activated gradually after arteriolar dilatation. Discrete nNOS neuron subsets may contribute either to the initiation or to the maintenance of the vascular response, suggesting a previously unappreciated temporal specificity to the role of NO in neurovascular coupling.
Collapse
Affiliation(s)
- Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Xue Y, Tang J, Zhang M, He Y, Fu J, Ding F. Durative sleep fragmentation with or without hypertension suppress rapid eye movement sleep and generate cerebrovascular dysfunction. Neurobiol Dis 2023:106222. [PMID: 37419254 DOI: 10.1016/j.nbd.2023.106222] [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: 01/02/2023] [Revised: 06/07/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023] Open
Abstract
Either hypertension or chronic insomnia is the risk factor of developing vascular dementia. Durative hypertension can induce vascular remodeling and is used for modeling small vessel disease in rodents. It remains undetermined if the combination of hypertension and sleep disturbance exacerbates vascular dysfunction or pathologies. Previously, we found chronic sleep fragmentation (SF) dampened cognition in young mice without disease predispositions. In the current study, we superimposed SF with hypertension modeling in young mice. Angiotensin II (AngII)-releasing osmotic mini pumps were subcutaneously implanted to generate persistent hypertension, while sham surgeries were performed as controls. Sleep fragmentation with repetitive arousals (10 s every 2 min) during light-on 12 h for consecutive 30 days, while mice undergoing normal sleep (NS) processes were set as controls. Sleep architectures, whisker-stimulated cerebral blood flow (CBF) changes, vascular responsiveness as well as vascular pathologies were compared among normal sleep plus sham (NS + sham), SF plus sham (SF + sham), normal sleep plus AngII (NS + AngII), and SF plus AngII (SF + AngII) groups. SF and hypertension both alter sleep structures, particularly suppressing REM sleep. SF no matter if combined with hypertension strongly suppressed whisker-stimulated CBF increase, suggesting the tight association with cognitive decline. Hypertension modeling sensitizes vascular responsiveness toward a vasoactive agent, Acetylcholine (ACh, 5 mg/ml, 10 μl) delivered via cisterna magna infusion, while SF exhibits a similar but much milder effect. None of the modeling above was sufficient to induce arterial or arteriole vascular remodeling, but SF or SF plus hypertension increased vascular network density constructed by all categories of cerebral vessels. The current study would potentially help understand the pathogenesis of vascular dementia, and the interconnection between sleep and vascular health.
Collapse
Affiliation(s)
- Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Miaoyi Zhang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Yifan He
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China.
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
11
|
Pluta R, Miziak B, Czuczwar SJ. Post-Ischemic Permeability of the Blood-Brain Barrier to Amyloid and Platelets as a Factor in the Maturation of Alzheimer's Disease-Type Brain Neurodegeneration. Int J Mol Sci 2023; 24:10739. [PMID: 37445917 DOI: 10.3390/ijms241310739] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this review is to present evidence of the impact of ischemic changes in the blood-brain barrier on the maturation of post-ischemic brain neurodegeneration with features of Alzheimer's disease. Understanding the processes involved in the permeability of the post-ischemic blood-brain barrier during recirculation will provide clinically relevant knowledge regarding the neuropathological changes that ultimately lead to dementia of the Alzheimer's disease type. In this review, we try to distinguish between primary and secondary neuropathological processes during and after ischemia. Therefore, we can observe two hit stages that contribute to Alzheimer's disease development. The onset of ischemic brain pathology includes primary ischemic neuronal damage and death followed by the ischemic injury of the blood-brain barrier with serum leakage of amyloid into the brain tissue, leading to increased ischemic neuronal susceptibility to amyloid neurotoxicity, culminating in the formation of amyloid plaques and ending in full-blown dementia of the Alzheimer's disease type.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
12
|
Zhukov O, He C, Soylu-Kucharz R, Cai C, Lauritzen AD, Aldana BI, Björkqvist M, Lauritzen M, Kucharz K. Preserved blood-brain barrier and neurovascular coupling in female 5xFAD model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1089005. [PMID: 37261266 PMCID: PMC10228387 DOI: 10.3389/fnagi.2023.1089005] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/17/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Dysfunction of the cerebral vasculature is considered one of the key components of Alzheimer's disease (AD), but the mechanisms affecting individual brain vessels are poorly understood. Methods Here, using in vivo two-photon microscopy in superficial cortical layers and ex vivo imaging across brain regions, we characterized blood-brain barrier (BBB) function and neurovascular coupling (NVC) at the level of individual brain vessels in adult female 5xFAD mice, an aggressive amyloid-β (Aβ) model of AD. Results We report a lack of abnormal increase in adsorptive-mediated transcytosis of albumin and preserved paracellular barrier for fibrinogen and small molecules despite an extensive load of Aβ. Likewise, the NVC responses to somatosensory stimulation were preserved at all regulatory segments of the microvasculature: penetrating arterioles, precapillary sphincters, and capillaries. Lastly, the Aβ plaques did not affect the density of capillary pericytes. Conclusion Our findings provide direct evidence of preserved microvascular function in the 5xFAD mice and highlight the critical dependence of the experimental outcomes on the choice of preclinical models of AD. We propose that the presence of parenchymal Aβ does not warrant BBB and NVC dysfunction and that the generalized view that microvascular impairment is inherent to Aβ aggregation may need to be revised.
Collapse
Affiliation(s)
- Oleg Zhukov
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chen He
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rana Soylu-Kucharz
- Biomarkers in Brain Disease, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Changsi Cai
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Martin Lauritzen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark
| | - Krzysztof Kucharz
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Uekawa K, Hattori Y, Ahn SJ, Seo J, Casey N, Anfray A, Zhou P, Luo W, Anrather J, Park L, Iadecola C. Border-associated macrophages promote cerebral amyloid angiopathy and cognitive impairment through vascular oxidative stress. RESEARCH SQUARE 2023:rs.3.rs-2719812. [PMID: 37162996 PMCID: PMC10168479 DOI: 10.21203/rs.3.rs-2719812/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background: Cerebral amyloid angiopathy (CAA) is a devastating condition common in patients with Alzheimer's disease but also observed in the general population. Vascular oxidative stress and neurovascular dysfunction have been implicated in CAA but the cellular source of reactive oxygen species (ROS) and related signaling mechanisms remain unclear. We tested the hypothesis that brain border-associated macrophages (BAM), yolk sac-derived myeloid cells closely apposed to parenchymal and leptomeningeal blood vessels, are the source of radicals through the Aβ-binding innate immunity receptor CD36, leading to neurovascular dysfunction, CAA, and cognitive impairment. Methods: Tg2576 mice and WT littermates were transplanted with CD36 -/- or CD36 +/+ bone marrow at 12-month of age and tested at 15 months. This approach enables the repopulation of perivascular and leptomeningeal compartments with CD36 -/- BAM. Neurovascular function was tested in anesthetized mice equipped with a cranial window in which cerebral blood flow was monitored by laser-Doppler flowmetry. Amyloid pathology and cognitive function were also examined. Results: The increase in blood flow evoked by whisker stimulation (functional hyperemia) or by endothelial and smooth muscle vasoactivity was markedly attenuated in WT®Tg2576 chimeras but was fully restored in CD36 -/- ®Tg2576 chimeras, in which BAM ROS production was suppressed. CAA-associated Aβ 1-40 , but not Aβ 1-42 , was reduced in CD36 -/- ®Tg2576 chimeras. Similarly, CAA, but not parenchymal plaques, was reduced in CD36 -/- ®Tg2576 chimeras. These beneficial vascular effects were associated with cognitive improvement. Finally, CD36 -/- mice were able to more efficiently clear exogenous Aβ 1-40 injected into the neocortex or the striatum. Conclusions: CD36 deletion in BAM suppresses ROS production and rescues the neurovascular dysfunction and damage induced by Aβ. CD36 deletion in BAM also reduced brain Aβ 1-40 and ameliorated CAA without affecting parenchyma plaques. Lack of CD36 enhanced the vascular clearance of exogenous Aβ. Restoration of neurovascular function and attenuation of CAA resulted in a near complete rescue of cognitive function. Collectively, these data implicate CNS BAM in the pathogenesis of CAA and raise the possibility that targeting BAM CD36 is beneficial in CAA and other conditions associated with vascular Aβ deposition and damage.
Collapse
|
14
|
Nabizadeh F, Balabandian M, Rostami MR, Mehrabi S, Sedighi M. Regional cerebral blood flow and brain atrophy in mild cognitive impairment and Alzheimer's disease. NEUROLOGY LETTERS 2023; 2:16-24. [PMID: 38327487 PMCID: PMC10849084 DOI: 10.52547/nl.2.1.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Objectives A decline in the regional cerebral blood flow (CBF) is proposed to be one of the initial changes in the Alzheimer's disease process. To date, there are limited data on the correlation between CBF decline and gray matter atrophy in mild cognitive impairment (MCI) and AD patients. to investigate the association between CBF with the gray matter structural parameters such as cortical volume, surface area, and thickness in AD, MCI, and healthy controls (HC). Methods Data from three groups of participants including 39 HC, 82 MCI, and 28 AD subjects were obtained from the Alzheimer's disease Neuroimaging Initiative (ADNI). One-way ANOVA and linear regression were used to compare data and find a correlation between structural parameters such as cortical volume, surface area, and thickness and CBF which measured by arterial spin labeling (ASL)-MRI. Results Our findings revealed a widespread significant correlation between the CBF and structural parameters in temporal, frontal, parietal, occipital, precentral gyrus, pericalcarine cortex, entorhinal cortex, supramarginal gyrus, fusiform, precuneus, and pallidum. Conclusion CBF decline may be a useful biomarker for MCI and AD and accurately reflect the structural changes related to AD. According to the present results, CBF decline, as measured by ASL-MRI, is correlated with lower measures of structural parameters in AD responsible regions. It means that CBF decline may reflect AD-associated atrophy across disease progression and is also used as an early biomarker for AD and MCI diagnosis.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Balabandian
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rostami
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soraya Mehrabi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mohsen Sedighi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
15
|
Crouzet C, Phan T, Wilson RH, Shin TJ, Choi B. Intrinsic, widefield optical imaging of hemodynamics in rodent models of Alzheimer's disease and neurological injury. NEUROPHOTONICS 2023; 10:020601. [PMID: 37143901 PMCID: PMC10152182 DOI: 10.1117/1.nph.10.2.020601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/30/2023] [Indexed: 05/06/2023]
Abstract
The complex cerebrovascular network is critical to controlling local cerebral blood flow (CBF) and maintaining brain homeostasis. Alzheimer's disease (AD) and neurological injury can result in impaired CBF regulation, blood-brain barrier breakdown, neurovascular dysregulation, and ultimately impaired brain homeostasis. Measuring cortical hemodynamic changes in rodents can help elucidate the complex physiological dynamics that occur in AD and neurological injury. Widefield optical imaging approaches can measure hemodynamic information, such as CBF and oxygenation. These measurements can be performed over fields of view that range from millimeters to centimeters and probe up to the first few millimeters of rodent brain tissue. We discuss the principles and applications of three widefield optical imaging approaches that can measure cerebral hemodynamics: (1) optical intrinsic signal imaging, (2) laser speckle imaging, and (3) spatial frequency domain imaging. Future work in advancing widefield optical imaging approaches and employing multimodal instrumentation can enrich hemodynamic information content and help elucidate cerebrovascular mechanisms that lead to the development of therapeutic agents for AD and neurological injury.
Collapse
Affiliation(s)
- Christian Crouzet
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Thinh Phan
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Biomedical Engineering, Irvine, California, United States
| | - Robert H. Wilson
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Medicine, Irvine, California, United States
| | - Teo Jeon Shin
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- Seoul National University, Department of Pediatric Dentistry and Dental Research Institute, Seoul, Republic of Korea
| | - Bernard Choi
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Biomedical Engineering, Irvine, California, United States
- University of California, Irvine, Department of Surgery, Irvine, California, United States
- University of California, Irvine, Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, California, United States
| |
Collapse
|
16
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
17
|
Ruiz-Uribe NE, Bracko O, Swallow M, Omurzakov A, Dash S, Uchida H, Xiang D, Haft-Javaherian M, Falkenhain K, Lamont ME, Ali M, Njiru BN, Chang HY, Tan AY, Xiang JZ, Iadecola C, Park L, Sanchez T, Nishimura N, Schaffer CB. Vascular oxidative stress causes neutrophil arrest in brain capillaries, leading to decreased cerebral blood flow and contributing to memory impairment in a mouse model of Alzheimer’s disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528710. [PMID: 36824768 PMCID: PMC9949082 DOI: 10.1101/2023.02.15.528710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION In this study, we explore the role of oxidative stress produced by NOX2-containing NADPH oxidase as a molecular mechanism causing capillary stalling and cerebral blood flow deficits in the APP/PS1 mouse model of AD. METHODS We inhibited NOX2 in APP/PS1 mice by administering a 10 mg/kg dose of the peptide inhibitor gp91-ds-tat i.p., for two weeks. We used in vivo two-photon imaging to measure capillary stalling, penetrating arteriole flow, and vascular inflammation. We also characterized short-term memory function and gene expression changes in cerebral microvessels. RESULTS We found that after NOX2 inhibition capillary stalling, as well as parenchymal and vascular inflammation, were significantly reduced. In addition, we found a significant increase in penetrating arteriole flow, followed by an improvement in short-term memory, and downregulation of inflammatory gene expression pathways. DISCUSSION Oxidative stress is a major mechanism leading to microvascular dysfunction in AD, and represents an important therapeutic target.
Collapse
|
18
|
Tayler HM, MacLachlan R, Güzel Ö, Miners JS, Love S. Elevated late-life blood pressure may maintain brain oxygenation and slow amyloid-β accumulation at the expense of cerebral vascular damage. Brain Commun 2023; 5:fcad112. [PMID: 37113314 PMCID: PMC10128877 DOI: 10.1093/braincomms/fcad112] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/16/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Hypertension in midlife contributes to cognitive decline and is a modifiable risk factor for dementia. The relationship between late-life hypertension and dementia is less clear. We have investigated the relationship of blood pressure and hypertensive status during late life (after 65 years) to post-mortem markers of Alzheimer's disease (amyloid-β and tau loads); arteriolosclerosis and cerebral amyloid angiopathy; and to biochemical measures of ante-mortem cerebral oxygenation (the myelin-associated glycoprotein:proteolipid protein-1 ratio, which is reduced in chronically hypoperfused brain tissue, and the level of vascular endothelial growth factor-A, which is upregulated by tissue hypoxia); blood-brain barrier damage (indicated by an increase in parenchymal fibrinogen); and pericyte content (platelet-derived growth factor receptor β, which declines with pericyte loss), in Alzheimer's disease (n = 75), vascular (n = 20) and mixed dementia (n = 31) cohorts. Systolic and diastolic blood pressure measurements were obtained retrospectively from clinical records. Non-amyloid small vessel disease and cerebral amyloid angiopathy were scored semiquantitatively. Amyloid-β and tau loads were assessed by field fraction measurement in immunolabelled sections of frontal and parietal lobes. Homogenates of frozen tissue from the contralateral frontal and parietal lobes (cortex and white matter) were used to measure markers of vascular function by enzyme-linked immunosorbent assay. Diastolic (but not systolic) blood pressure was associated with the preservation of cerebral oxygenation, correlating positively with the ratio of myelin-associated glycoprotein to proteolipid protein-1 and negatively with vascular endothelial growth factor-A in both the frontal and parietal cortices. Diastolic blood pressure correlated negatively with parenchymal amyloid-β in the parietal cortex. In dementia cases, elevated late-life diastolic blood pressure was associated with more severe arteriolosclerosis and cerebral amyloid angiopathy, and diastolic blood pressure correlated positively with parenchymal fibrinogen, indicating blood-brain barrier breakdown in both regions of the cortex. Systolic blood pressure was related to lower platelet-derived growth factor receptor β in controls in the frontal cortex and in dementia cases in the superficial white matter. We found no association between blood pressure and tau. Our findings demonstrate a complex relationship between late-life blood pressure, disease pathology and vascular function in dementia. We suggest that hypertension helps to reduce cerebral ischaemia (and may slow amyloid-β accumulation) in the face of increasing cerebral vascular resistance, but exacerbates vascular pathology.
Collapse
Affiliation(s)
- Hannah M Tayler
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Robert MacLachlan
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Özge Güzel
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - J Scott Miners
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Seth Love
- Correspondence to: Seth Love South West Dementia Brain Bank, University of Bristol Learning & Research Level 1, Southmead Hospital, Bristol, BS10 5NB, UK E-mail:
| |
Collapse
|
19
|
Hayes G, Pinto J, Sparks SN, Wang C, Suri S, Bulte DP. Vascular smooth muscle cell dysfunction in neurodegeneration. Front Neurosci 2022; 16:1010164. [PMID: 36440263 PMCID: PMC9684644 DOI: 10.3389/fnins.2022.1010164] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the key moderators of cerebrovascular dynamics in response to the brain's oxygen and nutrient demands. Crucially, VSMCs may provide a sensitive biomarker for neurodegenerative pathologies where vasculature is compromised. An increasing body of research suggests that VSMCs have remarkable plasticity and their pathophysiology may play a key role in the complex process of neurodegeneration. Furthermore, extrinsic risk factors, including environmental conditions and traumatic events can impact vascular function through changes in VSMC morphology. VSMC dysfunction can be characterised at the molecular level both preclinically, and clinically ex vivo. However the identification of VSMC dysfunction in living individuals is important to understand changes in vascular function at the onset and progression of neurological disorders such as dementia, Alzheimer's disease, and Parkinson's disease. A promising technique to identify changes in the state of cerebral smooth muscle is cerebrovascular reactivity (CVR) which reflects the intrinsic dynamic response of blood vessels in the brain to vasoactive stimuli in order to modulate regional cerebral blood flow (CBF). In this work, we review the role of VSMCs in the most common neurodegenerative disorders and identify physiological systems that may contribute to VSMC dysfunction. The evidence collected here identifies VSMC dysfunction as a strong candidate for novel therapeutics to combat the development and progression of neurodegeneration, and highlights the need for more research on the role of VSMCs and cerebrovascular dynamics in healthy and diseased states.
Collapse
Affiliation(s)
- Genevieve Hayes
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sierra N. Sparks
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Congxiyu Wang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Daniel P. Bulte
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Zheng L, Guo Y, Zhai X, Zhang Y, Chen W, Zhu Z, Xuan W, Li P. Perivascular macrophages in the CNS: From health to neurovascular diseases. CNS Neurosci Ther 2022; 28:1908-1920. [PMID: 36128654 PMCID: PMC9627394 DOI: 10.1111/cns.13954] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Brain perivascular macrophages (PVMs) are attracting increasing attention as this emerging cell population in the brain has multifaced roles in supporting the central nervous system structure, brain development, and maintaining physiological functions. They also widely participate in neurological diseases such as neurodegeneration and ischemic stroke. Moreover, PVMs have been reported to have both beneficial and detrimental effects under different pathological contexts. Advanced research technologies allowed the further in-depth study of PVMs and revealed novel concepts in their origins, differentiation, and regulatory mechanisms. Deepened understanding of the roles of PVMs in different brain pathological conditions can reveal novel phenotypic changes and regulatory signaling, which might pave the way for the development of novel treatment strategies targeting PVMs.
Collapse
Affiliation(s)
- Li Zheng
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunlu Guo
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaozhu Zhai
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ziyu Zhu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Xuan
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
21
|
Bojovic D, Stackhouse TL, Mishra A. Assaying activity-dependent arteriole and capillary responses in brain slices. NEUROPHOTONICS 2022; 9:031913. [PMID: 35558646 PMCID: PMC9089234 DOI: 10.1117/1.nph.9.3.031913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
Significance: Neurovascular coupling (NVC) is the process that increases cerebral blood flow in response to neuronal activity. NVC is orchestrated by signaling between neurons, glia, and vascular cells. Elucidating the mechanisms underlying NVC at different vascular segments and in different brain regions is imperative for understanding of brain function and mechanisms of dysfunction. Aim: Our goal is to describe a protocol for concurrently monitoring stimulation-evoked neuronal activity and resultant vascular responses in acute brain slices. Approach: We describe a step-by-step protocol that allows the study of endogenous NVC mechanisms engaged by neuronal activity in a controlled, reduced preparation. Results: This ex vivo NVC assay allows researchers to disentangle the mechanisms regulating the contractile responses of different vascular segments in response to neuronal firing independent of flow and pressure mediated effects from connected vessels. It also enables easy pharmacological manipulations in a simplified, reduced system and can be combined with Ca 2 + imaging or broader electrophysiology techniques to obtain multimodal data during NVC. Conclusions: The ex vivo NVC assay will facilitate investigations of cellular and molecular mechanisms that give rise to NVC and should serve as a valuable complement to in vivo imaging methods.
Collapse
Affiliation(s)
- Danica Bojovic
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
- Oregon Health & Science University, Vollum Institute, Portland, Oregon, United States
| | - Teresa L. Stackhouse
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
| | - Anusha Mishra
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
- Oregon Health & Science University, Knight Cardiovascular Institute, Portland, Oregon, United States
| |
Collapse
|
22
|
Gokcal E, Horn MJ, Becker JA, Das AS, Schwab K, Biffi A, Rost N, Rosand J, Viswanathan A, Polimeni JR, Johnson KA, Greenberg SM, Gurol ME. Effect of vascular amyloid on white matter disease is mediated by vascular dysfunction in cerebral amyloid angiopathy. J Cereb Blood Flow Metab 2022; 42:1272-1281. [PMID: 35086372 PMCID: PMC9207495 DOI: 10.1177/0271678x221076571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We postulated that vascular dysfunction mediates the relationship between amyloid load and white matter hyperintensities (WMH) in cerebral amyloid angiopathy (CAA). Thirty-eight cognitively healthy patients with CAA (mean age 70 ± 7.1) were evaluated. WMH was quantified and expressed as percent of total intracranial volume (pWMH) using structural MRI. Mean global cortical Distribution Volume Ratio representing Pittsburgh Compound B (PiB) uptake (PiB-DVR) was calculated from PET scans. Time-to-peak [TTP] of blood oxygen level-dependent response to visual stimulation was used as an fMRI measure of vascular dysfunction. Higher PiB-DVR correlated with prolonged TTP (r = 0.373, p = 0.021) and higher pWMH (r = 0.337, p = 0.039). Prolonged TTP also correlated with higher pWMH (r = 0.485, p = 0.002). In a multivariate linear regression model, TTP remained independently associated with pWMH (p = 0.006) while PiB-DVR did not (p = 0.225). In a bootstrapping model, TTP had a significant indirect effect (ab = 0.97, 95% CI: 0.137-2.461), supporting that the association between PiB-DVR and pWMH is mediated by TTP response. There was no longer a direct effect independent of the hypothesized pathway. Our study suggests that the effect of vascular amyloid load on white matter disease is mediated by vascular dysfunction in CAA. Amyloid lowering strategies might prevent pathophysiological processes leading to vascular dysfunction, therefore limiting ischemic brain injury.
Collapse
Affiliation(s)
- Elif Gokcal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mitchell J Horn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Alex Becker
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Alvin S Das
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandro Biffi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Natalia Rost
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Keith A Johnson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - M Edip Gurol
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Functionally linked potassium channel activity in cerebral endothelial and smooth muscle cells is compromised in Alzheimer's disease. Proc Natl Acad Sci U S A 2022; 119:e2204581119. [PMID: 35727988 PMCID: PMC9245656 DOI: 10.1073/pnas.2204581119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Patients with Alzheimer’s disease show hypoperfusion of the brain and this may contribute to disease progression. To elucidate underlying mechanisms, we studied pial arteries from 18-mo-old mice with Alzheimer’s disease due to overexpression of amyloid precursor protein. We found enhanced pressure-induced constriction of arteries because of reduction in ryanodine receptor-mediated, local calcium-release events (“Ca2+ sparks”) in arterial smooth muscle cells and a consequent decrease in the activity of large-conductance Ca2+-activated K+ (BK) channels. This phenotype was partially recapitulated by application of an amyloid-β peptide to healthy arteries. Our results will direct further research to restore cerebrovascular function, which is damaged in Alzheimer’s disease, leading to potentially new treatment options. The brain microcirculation is increasingly viewed as a potential target for disease-modifying drugs in the treatment of Alzheimer’s disease patients, reflecting a growing appreciation of evidence that cerebral blood flow is compromised in such patients. However, the pathogenic mechanisms in brain resistance arteries underlying blood flow defects have not yet been elucidated. Here we probed the roles of principal vasodilatory pathways in cerebral arteries using the APP23 mouse model of Alzheimer’s disease, in which amyloid precursor protein is increased approximately sevenfold, leading to neuritic plaques and cerebrovascular accumulation of amyloid-β similar to those in patients with Alzheimer’s disease. Pial arteries from APP23 mice (18 mo old) exhibited enhanced pressure-induced (myogenic) constriction because of a profound reduction in ryanodine receptor-mediated, local calcium-release events (“Ca2+ sparks”) in arterial smooth muscle cells and a consequent decrease in the activity of large-conductance Ca2+-activated K+ (BK) channels. The ability of the endothelial cell inward rectifier K+ (Kir2.1) channel to cause dilation was also compromised. Acute application of amyloid-β 1-40 peptide to cerebral arteries from wild-type mice partially recapitulated the BK dysfunction seen in APP23 mice but had no effect on Kir2.1 function. If mirrored in human Alzheimer’s disease, these tandem defects in K+ channel-mediated vasodilation could account for the clinical cerebrovascular presentation seen in patients: reduced blood flow and crippled functional hyperemia. These data direct future research toward approaches that reverse this dual vascular channel dysfunction, with the ultimate aim of restoring healthy cerebral blood flow and improving clinical outcomes.
Collapse
|
24
|
Vargas-George S, Dave KR. Models of cerebral amyloid angiopathy-related intracerebral hemorrhage. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
25
|
Reagan AM, Onos KD, Heuer SE, Sasner M, Howell GR. Improving mouse models for the study of Alzheimer's disease. Curr Top Dev Biol 2022; 148:79-113. [PMID: 35461569 DOI: 10.1016/bs.ctdb.2021.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease whose risk is influenced by genetic and environmental factors. Although a number of pathological hallmarks have been extensively studied over the last several decades, a complete picture of disease initiation and progression remains unclear. We now understand that numerous cell types and systems are involved in AD pathogenesis, and that this cellular profile may present differently for each individual, making the creation of relevant mouse models challenging. However, with increasingly diverse data made available by genome-wide association studies, we can identify and examine new genes and pathways involved in genetic risk for AD, many of which involve vascular health and inflammation. When developing mouse models, it is critical to assess (1) an aging timeline that represents onset and progression in humans, (2) genetic variants and context, (3) environmental factors present in human populations that result in both neuropathological and functional changes-themes that we address in this chapter.
Collapse
Affiliation(s)
| | | | - Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States.
| |
Collapse
|
26
|
Peters EC, Gee MT, Pawlowski LN, Kath AM, Polk FD, Vance CJ, Sacoman JL, Pires PW. Amyloid- β disrupts unitary calcium entry through endothelial NMDA receptors in mouse cerebral arteries. J Cereb Blood Flow Metab 2022; 42:145-161. [PMID: 34465229 PMCID: PMC8721780 DOI: 10.1177/0271678x211039592] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/07/2023]
Abstract
Transient increases in intracellular Ca2+ activate endothelium-dependent vasodilatory pathways. This process is impaired in cerebral amyloid angiopathy, where amyloid-β(1-40) accumulates around blood vessels. In neurons, amyloid-β impairs the Ca2+-permeable N-methyl-D-aspartate receptor (NMDAR), a mediator of endothelium-dependent dilation in arteries. We hypothesized that amyloid-β(1-40) reduces NMDAR-elicited Ca2+ signals in mouse cerebral artery endothelial cells, blunting dilation. Cerebral arteries isolated from 4-5 months-old, male and female cdh5:Gcamp8 mice were used for imaging of unitary Ca2+ influx through NMDAR (NMDAR sparklets) and intracellular Ca2+ transients. The NMDAR agonist NMDA (10 µmol/L) increased frequency of NMDAR sparklets and intracellular Ca2+ transients in endothelial cells; these effects were prevented by NMDAR antagonists D-AP5 and MK-801. Next, we tested if amyloid-β(1-40) impairs NMDAR-elicited Ca2+ transients. Cerebral arteries incubated with amyloid-β(1-40) (5 µmol/L) exhibited reduced NMDAR sparklets and intracellular Ca2+ transients. Lastly, we observed that NMDA-induced dilation of pial arteries is reduced by acute intraluminal amyloid-β(1-40), as well as in a mouse model of Alzheimer's disease, the 5x-FAD, linked to downregulation of Grin1 mRNA compared to wild-type littermates. These data suggest that endothelial NMDAR mediate dilation via Ca2+-dependent pathways, a process disrupted by amyloid-β(1-40) and impaired in 5x-FAD mice.
Collapse
Affiliation(s)
- Emily C Peters
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Michael T Gee
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Lukas N Pawlowski
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Allison M Kath
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Felipe D Polk
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Christopher J Vance
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Juliana L Sacoman
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| |
Collapse
|
27
|
Lourenço CF, Laranjinha J. Nitric Oxide Pathways in Neurovascular Coupling Under Normal and Stress Conditions in the Brain: Strategies to Rescue Aberrant Coupling and Improve Cerebral Blood Flow. Front Physiol 2021; 12:729201. [PMID: 34744769 PMCID: PMC8569710 DOI: 10.3389/fphys.2021.729201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023] Open
Abstract
The brain has impressive energy requirements and paradoxically, very limited energy reserves, implying its huge dependency on continuous blood supply. Aditionally, cerebral blood flow must be dynamically regulated to the areas of increased neuronal activity and thus, of increased metabolic demands. The coupling between neuronal activity and cerebral blood flow (CBF) is supported by a mechanism called neurovascular coupling (NVC). Among the several vasoactive molecules released by glutamatergic activation, nitric oxide (•NO) is recognized to be a key player in the process and essential for the development of the neurovascular response. Classically, •NO is produced in neurons upon the activation of the glutamatergic N-methyl-D-aspartate (NMDA) receptor by the neuronal isoform of nitric oxide synthase and promotes vasodilation by activating soluble guanylate cyclase in the smooth muscle cells of the adjacent arterioles. This pathway is part of a more complex network in which other molecular and cellular intervenients, as well as other sources of •NO, are involved. The elucidation of these interacting mechanisms is fundamental in understanding how the brain manages its energy requirements and how the failure of this process translates into neuronal dysfunction. Here, we aimed to provide an integrated and updated perspective of the role of •NO in the NVC, incorporating the most recent evidence that reinforces its central role in the process from both viewpoints, as a physiological mediator and a pathological stressor. First, we described the glutamate-NMDA receptor-nNOS axis as a central pathway in NVC, then we reviewed the link between the derailment of the NVC and neuronal dysfunction associated with neurodegeneration (with a focus on Alzheimer's disease). We further discussed the role of oxidative stress in the NVC dysfunction, specifically by decreasing the •NO bioavailability and diverting its bioactivity toward cytotoxicity. Finally, we highlighted some strategies targeting the rescue or maintenance of •NO bioavailability that could be explored to mitigate the NVC dysfunction associated with neurodegenerative conditions. In line with this, the potential modulatory effects of dietary nitrate and polyphenols on •NO-dependent NVC, in association with physical exercise, may be used as effective non-pharmacological strategies to promote the •NO bioavailability and to manage NVC dysfunction in neuropathological conditions.
Collapse
Affiliation(s)
- Cátia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
28
|
Hussain B, Fang C, Chang J. Blood-Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia. Front Neurosci 2021; 15:688090. [PMID: 34489623 PMCID: PMC8418300 DOI: 10.3389/fnins.2021.688090] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) plays a vital role in maintaining the specialized microenvironment of the neural tissue. It separates the peripheral circulatory system from the brain parenchyma while facilitating communication. Alterations in the distinct physiological properties of the BBB lead to BBB breakdown associated with normal aging and various neurodegenerative diseases. In this review, we first briefly discuss the aging process, then review the phenotypes and mechanisms of BBB breakdown associated with normal aging that further cause neurodegeneration and cognitive impairments. We also summarize dementia such as Alzheimer's disease (AD) and vascular dementia (VaD) and subsequently discuss the phenotypes and mechanisms of BBB disruption in dementia correlated with cognition decline. Overlaps between AD and VaD are also discussed. Techniques that could identify biomarkers associated with BBB breakdown are briefly summarized. Finally, we concluded that BBB breakdown could be used as an emerging biomarker to assist to diagnose cognitive impairment associated with normal aging and dementia.
Collapse
Affiliation(s)
- Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
29
|
Quintana DD, Anantula Y, Garcia JA, Engler-Chiurazzi EB, Sarkar SN, Corbin DR, Brown CM, Simpkins JW. Microvascular degeneration occurs before plaque onset and progresses with age in 3xTg AD mice. Neurobiol Aging 2021; 105:115-128. [PMID: 34062487 PMCID: PMC9703920 DOI: 10.1016/j.neurobiolaging.2021.04.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/06/2023]
Abstract
Heart disease and vascular disease positively correlate with the incidence of Alzheimer's disease (AD). Although there is ostensible involvement of dysfunctional cerebrovasculature in AD pathophysiology, the characterization of the specific changes and development of vascular injury during AD remains unclear. In the present study, we established a time-course for the structural changes and degeneration of the angioarchitecture in AD. We used cerebrovascular corrosion cast and µCT imaging to evaluate the geometry, topology, and complexity of the angioarchitecture in the brain of wild type and 3xTg AD mice. We hypothesized that changes to the microvasculature occur early during the disease, and these early identifiable aberrations would be more prominent in the brain subregions implicated in the cognitive decline of AD. Whole-brain analysis of the angioarchitecture indicated early morphological abnormalities and degeneration of microvascular networks in 3xTg AD mice. Our analysis of the hippocampus and cortical subregions revealed microvascular degeneration with onset and progression that was subregion dependent.
Collapse
Affiliation(s)
- Dominic D Quintana
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Yamini Anantula
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Jorge A Garcia
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Elizabeth B Engler-Chiurazzi
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Saumyendra N Sarkar
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Deborah R Corbin
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Candice M Brown
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - James W Simpkins
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV.
| |
Collapse
|
30
|
Stackhouse TL, Mishra A. Neurovascular Coupling in Development and Disease: Focus on Astrocytes. Front Cell Dev Biol 2021; 9:702832. [PMID: 34327206 PMCID: PMC8313501 DOI: 10.3389/fcell.2021.702832] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Neurovascular coupling is a crucial mechanism that matches the high energy demand of the brain with a supply of energy substrates from the blood. Signaling within the neurovascular unit is responsible for activity-dependent changes in cerebral blood flow. The strength and reliability of neurovascular coupling form the basis of non-invasive human neuroimaging techniques, including blood oxygen level dependent (BOLD) functional magnetic resonance imaging. Interestingly, BOLD signals are negative in infants, indicating a mismatch between metabolism and blood flow upon neural activation; this response is the opposite of that observed in healthy adults where activity evokes a large oversupply of blood flow. Negative neurovascular coupling has also been observed in rodents at early postnatal stages, further implying that this is a process that matures during development. This rationale is consistent with the morphological maturation of the neurovascular unit, which occurs over a similar time frame. While neurons differentiate before birth, astrocytes differentiate postnatally in rodents and the maturation of their complex morphology during the first few weeks of life links them with synapses and the vasculature. The vascular network is also incomplete in neonates and matures in parallel with astrocytes. Here, we review the timeline of the structural maturation of the neurovascular unit with special emphasis on astrocytes and the vascular tree and what it implies for functional maturation of neurovascular coupling. We also discuss similarities between immature astrocytes during development and reactive astrocytes in disease, which are relevant to neurovascular coupling. Finally, we close by pointing out current gaps in knowledge that must be addressed to fully elucidate the mechanisms underlying neurovascular coupling maturation, with the expectation that this may also clarify astrocyte-dependent mechanisms of cerebrovascular impairment in neurodegenerative conditions in which reduced or negative neurovascular coupling is noted, such as stroke and Alzheimer’s disease.
Collapse
Affiliation(s)
- Teresa L Stackhouse
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States.,Knight Cardiovascular Institute, Oregon Health & Sciences University, Portland, OR, United States
| |
Collapse
|
31
|
Bracko O, Cruz Hernández JC, Park L, Nishimura N, Schaffer CB. Causes and consequences of baseline cerebral blood flow reductions in Alzheimer's disease. J Cereb Blood Flow Metab 2021; 41:1501-1516. [PMID: 33444096 PMCID: PMC8221770 DOI: 10.1177/0271678x20982383] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
Reductions of baseline cerebral blood flow (CBF) of ∼10-20% are a common symptom of Alzheimer's disease (AD) that appear early in disease progression and correlate with the severity of cognitive impairment. These CBF deficits are replicated in mouse models of AD and recent work shows that increasing baseline CBF can rapidly improve the performance of AD mice on short term memory tasks. Despite the potential role these data suggest for CBF reductions in causing cognitive symptoms and contributing to brain pathology in AD, there remains a poor understanding of the molecular and cellular mechanisms causing them. This review compiles data on CBF reductions and on the correlation of AD-related CBF deficits with disease comorbidities (e.g. cardiovascular and genetic risk factors) and outcomes (e.g. cognitive performance and brain pathology) from studies in both patients and mouse models, and discusses several potential mechanisms proposed to contribute to CBF reductions, based primarily on work in AD mouse models. Future research aimed at improving our understanding of the importance of and interplay between different mechanisms for CBF reduction, as well as at determining the role these mechanisms play in AD patients could guide the development of future therapies that target CBF reductions in AD.
Collapse
Affiliation(s)
- Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jean C Cruz Hernández
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
32
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
33
|
She M, Shang S, Hu N, Chen C, Dang L, Gao L, Wei S, Huo K, Wang J, Wang J, Qu Q. Blood Pressure Level Is Associated With Changes in Plasma Aβ 1 -40 and Aβ 1-42 Levels: A Cross-sectional Study Conducted in the Suburbs of Xi'an, China. Front Aging Neurosci 2021; 13:650679. [PMID: 34149395 PMCID: PMC8211897 DOI: 10.3389/fnagi.2021.650679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/06/2021] [Indexed: 11/23/2022] Open
Abstract
Objectives: Amyloid-β (Aβ) deposition in the brain is the hallmark of Alzheimer’s disease (AD) pathology. Hypertension is a risk factor for AD, but the effects of hypertension on Aβ deposition are not fully determined. Considering peripheral Aβ closely relates to Aβ deposition in the brain, we investigated the relationships between blood pressure (BP) level and plasma Aβ concentrations. Methods: One-thousand and sixty-nine participants (age above 45) from a village in the suburbs of Xi’an, China were enrolled. Questionnaires and validated Chinese versions of the Mini-Mental State Examination (MMSE) were used to collect information about vascular risk factors and assess cognition function. The apolipoprotein E (ApoE) genotype was detected using PCR and sequencing. Plasma Aβ levels were measured using ELISA. The associations between BP and plasma Aβ levels were analyzed by using multivariate linear regression. Results: Plasma Aβ1–40 level was higher in high BP group than that in normal BP group (53.34 ± 8.50 pg/ml vs. 51.98 ± 8.96 pg/ml, P = 0.013), in high SBP group than that in normal SBP group (53.68 ± 8.69 pg/ml vs. 51.88 ± 8.80 pg/ml, P = 0.001) and in high MABP group than that in normal MABP group (54.05 ± 8.78 pg/ml vs. 52.04 ± 8.75 pg/ml, P = 0.001). After controlling for the confounding factors, SBP (b = 0.078, P < 0.001), DBP (b = 0.090, P = 0.008) and MABP (b = 0.104, P < 0.001) correlated with plasma Aβ1–40 level positively in ApoE ε4 non-carriers, but not ApoE ε4 carriers. Conclusions: Elevated BP levels were associated with increased plasma Aβ1–40 levels in middle-aged and elderly ApoE ε4 non-carriers.
Collapse
Affiliation(s)
- Meilin She
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Neurology, Yulin Hospital of Traditional Chinese Medicine, Shaanxi, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ningwei Hu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Huyi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Palmer JC, Tayler HM, Dyer L, Kehoe PG, Paton JFR, Love S. Zibotentan, an Endothelin A Receptor Antagonist, Prevents Amyloid-β-Induced Hypertension and Maintains Cerebral Perfusion. J Alzheimers Dis 2021; 73:1185-1199. [PMID: 31903990 PMCID: PMC7081103 DOI: 10.3233/jad-190630] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cerebral blood flow is reduced in Alzheimer’s disease (AD), which is associated with mid-life hypertension. In people with increased cerebral vascular resistance due to vertebral artery or posterior communicating artery hypoplasia, there is evidence that hypertension develops as a protective mechanism to maintain cerebral perfusion. In AD, amyloid-β (Aβ) accumulation may similarly raise cerebral vascular resistance by upregulation of the cerebral endothelin system. The level of endothelin-1 in brain tissue correlates positively with Aβ load and negatively with markers of cerebral hypoperfusion such as increased vascular endothelial growth factor. We previously showed that cerebroventricular infusion of Aβ40 exacerbated pre-existing hypertension in Dahl rats. We have investigated the effects of 28-day cerebral infusion of Aβ40 on blood pressure and heart rate and their variability; carotid flow; endothelin-1; and markers of cerebral oxygenation, in the (normotensive) Wistar rat, and the modulatory influence of the endothelin A receptor antagonist Zibotentan (ZD4054). Cerebral infusion of Aβ caused progressive rise in blood pressure (p < 0.0001) (paired t-test: increase of 3 (0.1–5.6) mmHg (p = 0.040)), with evidence of reduced baroreflex responsiveness, and accumulation of Aβ and elevated endothelin-1 in the vicinity of the infusion. Oral Zibotentan (3 mg/kg/d, administered for 31 d) abrogated the effects of Aβ40 infusion on baroreflex responsiveness and blood pressure, which declined, although without reduction in carotid blood flow, and Zibotentan caused uncoupling of the positive linear relationship between endothelin-1 and vascular endothelial growth factor, which as a sensor of tissue oxygenation would be expected to increase if there were hypoperfusion.
Collapse
Affiliation(s)
- Jennifer C Palmer
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Hannah M Tayler
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Laurence Dyer
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Patrick G Kehoe
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, New Zealand
| | - Seth Love
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
35
|
Biomaterials in treatment of Alzheimer's disease. Neurochem Int 2021; 145:105008. [PMID: 33684545 DOI: 10.1016/j.neuint.2021.105008] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a non-recoverable progressive neurodegenerative disorder most prevalent but not limited to the old age population. After all the scientific efforts, there are still many unmet criteria and loopholes in available treatment and diagnostic strategies, limiting their efficacy. The poor drug efficacy is attributed to various biological hurdles, including blood-brain barrier (BBB) and peripheral side effects as most prominent ones and the lack of promising carriers to precisely deliver the drug to the brain by conserving its therapeutic potency. The increasing disease prevalence and unavailability of effective therapy calls for developing a more innovative, convenient and affordable way to treat AD. To fulfill such need, researchers explored various biomaterials to develop potential vectors or other forms to target the bioactives in the brain by preserving their inherent properties, improving the existing lacuna like poor solubility, permeability and bioavailability etc. and minimize the side effect. The unique characteristic properties of biomaterials are used to develop different drug carriers, surface modifying target active ligands, functional carriers, drug conjugate, biosensing probe, diagnostic tool and many more. The nanoparticulate system and other colloidal carriers like hydrogel and biodegradable scaffold can effectively target the drug moieties to the brain. Also, the use of different target-acting ligands and stimuli-responsive carriers assures the site-specificity and controlled release at the desired site by interaction with receptors and various exo- and endogenous stimuli. This review article has highlighted the application of biomaterials for targeting the drug to the brain and as promising diagnostic tools to detect the markers for better AD management. The work particularly focuses on the use of biomaterials as smart drug carriers including pH, thermo, photo, electro and magnetically triggered system; novel drug carriers for brain targeting including polymeric carriers (polymeric nanoparticle, dendrimer and polymeric micelle); lipid carrier (liposome, nanoemulsion, NLC and SLN); inorganic nanoparticles (quantum dots, gold nanoparticles etc.); and other drug vectors (hydrogel, biodegradable scaffold, and carbon nanotube) in treatment of AD. It also highlighted the application of some novel carrier systems and biomaterials as biosensor and other diagnostic tools for early and precise AD diagnosis.
Collapse
|
36
|
Mughal A, Harraz OF, Gonzales AL, Hill-Eubanks D, Nelson MT. PIP 2 Improves Cerebral Blood Flow in a Mouse Model of Alzheimer's Disease. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab010. [PMID: 33763649 PMCID: PMC7955025 DOI: 10.1093/function/zqab010] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia and a substantial healthcare burden. Despite this, few treatment options are available for controlling AD symptoms. Notably, neuronal activity-dependent increases in cortical cerebral blood flow (CBF; functional hyperemia) are attenuated in AD patients, but the associated pathological mechanisms are not fully understood at the molecular level. A fundamental mechanism underlying functional hyperemia is activation of capillary endothelial inward-rectifying K+ (Kir2.1) channels by neuronally derived potassium (K+), which evokes a retrograde capillary-to-arteriole electrical signal that dilates upstream arterioles, increasing blood delivery to downstream active regions. Here, using a mouse model of familial AD (5xFAD), we tested whether this impairment in functional hyperemia is attributable to reduced activity of capillary Kir2.1 channels. In vivo CBF measurements revealed significant reductions in whisker stimulation (WS)-induced and K+-induced hyperemic responses in 5xFAD mice compared with age-matched controls. Notably, measurements of whole-cell currents in freshly isolated 5xFAD capillary endothelial cells showed that Kir2.1 current density was profoundly reduced, suggesting a defect in Kir2.1 function. Because Kir2.1 activity absolutely depends on binding of phosphatidylinositol 4,5-bisphosphate (PIP2) to the channel, we hypothesized that capillary Kir2.1 channel impairment could be corrected by exogenously supplying PIP2. As predicted, a PIP2 analog restored Kir2.1 current density to control levels. More importantly, systemic administration of PIP2 restored K+-induced CBF increases and WS-induced functional hyperemic responses in 5xFAD mice. Collectively, these data provide evidence that PIP2-mediated restoration of capillary endothelial Kir2.1 function improves neurovascular coupling and CBF in the setting of AD.
Collapse
Affiliation(s)
- Amreen Mughal
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Albert L Gonzales
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - David Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Mark T Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA,Division of Cardiovascular Sciences, University of Manchester, Manchester, UK,Address correspondence to M.T.N. (e-mail: )
| |
Collapse
|
37
|
Alzheimer's Disease and Vascular Aging: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:942-951. [PMID: 32130930 PMCID: PMC8046164 DOI: 10.1016/j.jacc.2019.10.062] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/27/2019] [Indexed: 01/23/2023]
Abstract
Alzheimer’s disease, the leading cause of dementia in the elderly, is a neurodegenerative condition characterized by accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, age-related vascular changes accompany or even precede the development of Alzheimer’s pathology, raising the possibility that they may have a pathogenic role. This review provides an appraisal of the alterations in cerebral and systemic vasculature, the heart, and hemostasis that occur in Alzheimer’s disease and their relationships to cognitive impairment. Although the molecular pathogenesis of these alterations remains to be defined, amyloid-β is a likely contributor in the brain as in the heart. Collectively, the evidence suggests that vascular pathology is a likely pathogenic contributor to age-related dementia, including Alzheimer’s disease, inextricably linked to disease onset and progression. Consequently, the contribution of vascular factors should be considered in preventive, diagnostic, and therapeutic approaches to address one of the major health challenges of our time.
Collapse
|
38
|
Stakos DA, Stamatelopoulos K, Bampatsias D, Sachse M, Zormpas E, Vlachogiannis NI, Tual-Chalot S, Stellos K. The Alzheimer's Disease Amyloid-Beta Hypothesis in Cardiovascular Aging and Disease: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:952-967. [PMID: 32130931 PMCID: PMC7042886 DOI: 10.1016/j.jacc.2019.12.033] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Aging-related cellular and molecular processes including low-grade inflammation are major players in the pathogenesis of cardiovascular disease (CVD) and Alzheimer's disease (AD). Epidemiological studies report an independent interaction between the development of dementia and the incidence of CVD in several populations, suggesting the presence of overlapping molecular mechanisms. Accumulating experimental and clinical evidence suggests that amyloid-beta (Aβ) peptides may function as a link among aging, CVD, and AD. Aging-related vascular and cardiac deposition of Αβ induces tissue inflammation and organ dysfunction, both important components of the Alzheimer's disease amyloid hypothesis. In this review, the authors describe the determinants of Aβ metabolism, summarize the effects of Aβ on atherothrombosis and cardiac dysfunction, discuss the clinical value of Αβ1-40 in CVD prognosis and patient risk stratification, and present the therapeutic interventions that may alter Aβ metabolism in humans.
Collapse
Affiliation(s)
- Dimitrios A Stakos
- Cardiology Department, Democritus University of Thrace, Alexandroupolis, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece; Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Marco Sachse
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Medical School, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Eleftherios Zormpas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nikolaos I Vlachogiannis
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon Tual-Chalot
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Konstantinos Stellos
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
39
|
Korte N, Nortley R, Attwell D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer's disease. Acta Neuropathol 2020; 140:793-810. [PMID: 32865691 PMCID: PMC7666276 DOI: 10.1007/s00401-020-02215-w] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/15/2020] [Accepted: 08/15/2020] [Indexed: 02/08/2023]
Abstract
Therapies targeting late events in Alzheimer's disease (AD), including aggregation of amyloid beta (Aβ) and hyperphosphorylated tau, have largely failed, probably because they are given after significant neuronal damage has occurred. Biomarkers suggest that the earliest event in AD is a decrease of cerebral blood flow (CBF). This is caused by constriction of capillaries by contractile pericytes, probably evoked by oligomeric Aβ. CBF is also reduced by neutrophil trapping in capillaries and clot formation, perhaps secondary to the capillary constriction. The fall in CBF potentiates neurodegeneration by upregulating the BACE1 enzyme that makes Aβ and by promoting tau hyperphosphorylation. Surprisingly, therefore, CBF reduction may play a crucial role in driving cognitive decline by initiating the amyloid cascade itself, or being caused by and amplifying Aβ production. Here, we review developments in this area that are neglected in current approaches to AD, with the aim of promoting novel mechanism-based therapeutic approaches.
Collapse
Affiliation(s)
- Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ross Nortley
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
40
|
tPA Deficiency Underlies Neurovascular Coupling Dysfunction by Amyloid-β. J Neurosci 2020; 40:8160-8173. [PMID: 32928888 DOI: 10.1523/jneurosci.1140-20.2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/29/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
The amyloid-β (Aβ) peptide, a key pathogenic factor in Alzheimer's disease, attenuates the increase in cerebral blood flow (CBF) evoked by neural activity (functional hyperemia), a vital homeostatic response in which NMDA receptors (NMDARs) play a role through nitric oxide, and the CBF increase produced by endothelial factors. Tissue plasminogen activator (tPA), which is reduced in Alzheimer's disease and in mouse models of Aβ accumulation, is required for the full expression of the NMDAR-dependent component of functional hyperemia. Therefore, we investigated whether tPA is involved in the neurovascular dysfunction of Aβ. tPA activity was reduced, and the tPA inhibitor plasminogen inhibitor-1 (PAI-1) was increased in male mice expressing the Swedish mutation of the amyloid precursor protein (tg2576). Counteracting the tPA reduction with exogenous tPA or with pharmacological inhibition or genetic deletion of PAI-1 completely reversed the attenuation of the CBF increase evoked by whisker stimulation but did not ameliorate the response to the endothelium-dependent vasodilator acetylcholine. The tPA deficit attenuated functional hyperemia by suppressing NMDAR-dependent nitric oxide production during neural activity. Pharmacological inhibition of PAI-1 increased tPA activity, prevented neurovascular uncoupling, and ameliorated cognition in 11- to 12-month-old tg2576 mice, effects associated with a reduction of cerebral amyloid angiopathy but not amyloid plaques. The data unveil a selective role of the tPA in the suppression of functional hyperemia induced by Aβ and in the mechanisms of cerebral amyloid angiopathy, and support the possibility that modulation of the PAI-1-tPA pathway may be beneficial in diseases associated with amyloid accumulation.SIGNIFICANCE STATEMENT Amyloid-β (Aβ) peptides have profound neurovascular effects that may contribute to cognitive impairment in Alzheimer's disease. We found that Aβ attenuates the increases in blood flow evoked by neural activation through a reduction in tissue plasminogen activator (tPA) caused by upregulation of its endogenous inhibitor plasminogen inhibitor-1 (PAI-1). tPA deficiency prevents NMDA receptors from triggering nitric oxide production, thereby attenuating the flow increase evoked by neural activity. PAI-1 inhibition restores tPA activity, rescues neurovascular coupling, reduces amyloid deposition around blood vessels, and improves cognition in a mouse model of Aβ accumulation. The findings demonstrate a previously unappreciated role of tPA in Aβ-related neurovascular dysfunction and in vascular amyloid deposition. Restoration of tPA activity could be of therapeutic value in diseases associated with amyloid accumulation.
Collapse
|
41
|
Medin aggregation causes cerebrovascular dysfunction in aging wild-type mice. Proc Natl Acad Sci U S A 2020; 117:23925-23931. [PMID: 32900929 DOI: 10.1073/pnas.2011133117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Medin is the most common amyloid known in humans, as it can be found in blood vessels of the upper body in virtually everybody over 50 years of age. However, it remains unknown whether deposition of Medin plays a causal role in age-related vascular dysfunction. We now report that aggregates of Medin also develop in the aorta and brain vasculature of wild-type mice in an age-dependent manner. Strikingly, genetic deficiency of the Medin precursor protein, MFG-E8, eliminates not only vascular aggregates but also prevents age-associated decline of cerebrovascular function in mice. Given the prevalence of Medin aggregates in the general population and its role in vascular dysfunction with aging, targeting Medin may become a novel approach to sustain healthy aging.
Collapse
|
42
|
De la Rosa A, Olaso-Gonzalez G, Arc-Chagnaud C, Millan F, Salvador-Pascual A, García-Lucerga C, Blasco-Lafarga C, Garcia-Dominguez E, Carretero A, Correas AG, Viña J, Gomez-Cabrera MC. Physical exercise in the prevention and treatment of Alzheimer's disease. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:394-404. [PMID: 32780691 PMCID: PMC7498620 DOI: 10.1016/j.jshs.2020.01.004] [Citation(s) in RCA: 249] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/26/2019] [Accepted: 12/12/2019] [Indexed: 05/02/2023]
Abstract
Dementia is one of the greatest global challenges for health and social care in the 21st century. Alzheimer's disease (AD), the most common type of dementia, is by no means an inevitable consequence of growing old. Several lifestyle factors may increase, or reduce, an individual's risk of developing AD. Much has been written over the ages about the benefits of exercise and physical activity. Among the risk factors associated with AD is a low level of physical activity. The relationship between physical and mental health was established several years ago. In this review, we discuss the role of exercise (aerobic and resistance) training as a therapeutic strategy for the treatment and prevention of AD. Older adults who exercise are more likely to maintain cognition. We address the main protective mechanism on brain function modulated by physical exercise by examining both human and animal studies. We will pay especial attention to the potential role of exercise in the modulation of amyloid β turnover, inflammation, synthesis and release of neurotrophins, and improvements in cerebral blood flow. Promoting changes in lifestyle in presymptomatic and predementia disease stages may have the potential for delaying one-third of dementias worldwide. Multimodal interventions that include the adoption of an active lifestyle should be recommended for older populations.
Collapse
Affiliation(s)
- Adrian De la Rosa
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Gloria Olaso-Gonzalez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Coralie Arc-Chagnaud
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain; INRA, UMR866 Muscle dynamics and metabolism, University of Montpellier, F-34060, Montpellier, France
| | - Fernando Millan
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Andrea Salvador-Pascual
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | | | | | - Esther Garcia-Dominguez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Aitor Carretero
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Angela G Correas
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain
| | - Jose Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain.
| | - Mari Carmen Gomez-Cabrera
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Insitute of Health Research-INCLIVA, Valencia 46010, Spain.
| |
Collapse
|
43
|
The Role of Neurovascular System in Neurodegenerative Diseases. Mol Neurobiol 2020; 57:4373-4393. [PMID: 32725516 DOI: 10.1007/s12035-020-02023-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
The neurovascular system (NVS), which consisted of neurons, glia, and vascular cells, is a functional and structural unit of the brain. The NVS regulates blood-brain barrier (BBB) permeability and cerebral blood flow (CBF), thereby maintaining the brain's microenvironment for normal functioning, neuronal survival, and information processing. Recent studies have highlighted the role of vascular dysfunction in several neurodegenerative diseases. This is not unexpected since both nervous and vascular systems are functionally interdependent and show close anatomical apposition, as well as similar molecular pathways. However, despite extensive research, the precise mechanism by which neurovascular dysfunction contributes to neurodegeneration remains incomplete. Therefore, understanding the mechanisms of neurovascular dysfunction in disease conditions may allow us to develop potent and effective therapies for prevention and treatment of neurodegenerative diseases. This review article summarizes the current research in the context of neurovascular signaling associated with neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). We also discuss the potential implication of neurovascular factor as a novel therapeutic target and prognostic marker in patients with neurodegenerative conditions. Graphical Abstract.
Collapse
|
44
|
Gatti L, Tinelli F, Scelzo E, Arioli F, Di Fede G, Obici L, Pantoni L, Giaccone G, Caroppo P, Parati EA, Bersano A. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int J Mol Sci 2020; 21:ijms21103435. [PMID: 32414028 PMCID: PMC7279405 DOI: 10.3390/ijms21103435] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), one of the main types of cerebral small vessel disease, is a major cause of spontaneous intracerebral haemorrhage and an important contributor to cognitive decline in elderly patients. Despite the number of experimental in vitro studies and animal models, the pathophysiology of CAA is still largely unknown. Although several pathogenic mechanisms including an unbalance between production and clearance of amyloid beta (Aβ) protein as well as ‘the prion hypothesis’ have been invoked as possible disease triggers, they do not explain completely the disease pathogenesis. This incomplete disease knowledge limits the implementation of treatments able to prevent or halt the clinical progression. The continuous increase of CAA patients makes imperative the development of suitable experimental in vitro or animal models to identify disease biomarkers and new pharmacological treatments that could be administered in the early disease stages to prevent irreversible changes and disease progression.
Collapse
Affiliation(s)
- Laura Gatti
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Francesca Tinelli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Emma Scelzo
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Francesco Arioli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Giuseppe Di Fede
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Laura Obici
- Amyloidosis Research and Treatment Centre, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Leonardo Pantoni
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Giorgio Giaccone
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Paola Caroppo
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Eugenio Agostino Parati
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
- Correspondence: ; Tel.: +39-0223943310
| |
Collapse
|
45
|
Caballero MÁA, Song Z, Rubinski A, Duering M, Dichgans M, Park DC, Ewers M. Age‐dependent amyloid deposition is associated with white matter alterations in cognitively normal adults during the adult life span. Alzheimers Dement 2020; 16:651-661. [DOI: 10.1002/alz.12062] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 12/10/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Affiliation(s)
| | - Zhuang Song
- Center for Vital LongevityUniversity of Texas at Dallas Dallas Texas
| | - Anna Rubinski
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| | - Marco Duering
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich Germany
| | - Denise C. Park
- Center for Vital LongevityUniversity of Texas at Dallas Dallas Texas
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| |
Collapse
|
46
|
Potential Therapeutic Approaches for Cerebral Amyloid Angiopathy and Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21061992. [PMID: 32183348 PMCID: PMC7139812 DOI: 10.3390/ijms21061992] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a cerebrovascular disease directly implicated in Alzheimer’s disease (AD) pathogenesis through amyloid-β (Aβ) deposition, which may cause the development and progression of dementia. Despite extensive studies to explore drugs targeting Aβ, clinical benefits have not been reported in large clinical trials in AD patients or presymptomatic individuals at a risk for AD. However, recent studies on CAA and AD have provided novel insights regarding CAA- and AD-related pathogenesis. This work has revealed potential therapeutic targets, including Aβ drainage pathways, Aβ aggregation, oxidative stress, and neuroinflammation. The functional significance and therapeutic potential of bioactive molecules such as cilostazol and taxifolin have also become increasingly evident. Furthermore, recent epidemiological studies have demonstrated that serum levels of a soluble form of triggering receptor expressed on myeloid cells 2 (TREM2) may have clinical significance as a potential novel predictive biomarker for dementia incidence. This review summarizes recent advances in CAA and AD research with a focus on discussing future research directions regarding novel therapeutic approaches and predictive biomarkers for CAA and AD.
Collapse
|
47
|
Leira Y, Carballo Á, Orlandi M, Aldrey JM, Pías‐Peleteiro JM, Moreno F, Vázquez‐Vázquez L, Campos F, D’Aiuto F, Castillo J, Sobrino T, Blanco J. Periodontitis and systemic markers of neurodegeneration: A case–control study. J Clin Periodontol 2020; 47:561-571. [DOI: 10.1111/jcpe.13267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/28/2019] [Accepted: 02/03/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Yago Leira
- Periodontology Unit UCL Eastman Dental Institute and NIHR UCLH Biomedical Research Centre University College London London UK
- Periodontology Unit Faculty of Medicine and Odontology University of Santiago de Compostela Santiago de Compostela Spain
- Medical‐Surgical Dentistry (OMEQUI) Research Group Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Álvaro Carballo
- Periodontology Unit Faculty of Medicine and Odontology University of Santiago de Compostela Santiago de Compostela Spain
| | - Marco Orlandi
- Periodontology Unit UCL Eastman Dental Institute and NIHR UCLH Biomedical Research Centre University College London London UK
| | - José Manuel Aldrey
- Dementia Unit Department of Neurology Clinical University Hospital Santiago de Compostela Spain
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Juan Manuel Pías‐Peleteiro
- Dementia Unit Department of Neurology Clinical University Hospital Santiago de Compostela Spain
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Federico Moreno
- Periodontology Unit UCL Eastman Dental Institute and NIHR UCLH Biomedical Research Centre University College London London UK
| | - Laura Vázquez‐Vázquez
- Dementia Unit Department of Neurology Clinical University Hospital Santiago de Compostela Spain
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Francesco D’Aiuto
- Periodontology Unit UCL Eastman Dental Institute and NIHR UCLH Biomedical Research Centre University College London London UK
| | - José Castillo
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Juan Blanco
- Periodontology Unit Faculty of Medicine and Odontology University of Santiago de Compostela Santiago de Compostela Spain
- Medical‐Surgical Dentistry (OMEQUI) Research Group Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| |
Collapse
|
48
|
van Veluw SJ, Hou SS, Calvo-Rodriguez M, Arbel-Ornath M, Snyder AC, Frosch MP, Greenberg SM, Bacskai BJ. Vasomotion as a Driving Force for Paravascular Clearance in the Awake Mouse Brain. Neuron 2020; 105:549-561.e5. [PMID: 31810839 PMCID: PMC7028316 DOI: 10.1016/j.neuron.2019.10.033] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/09/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022]
Abstract
Paravascular drainage of solutes, including β-amyloid (Aβ), appears to be an important process in brain health and diseases such as Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). However, the major driving force for clearance remains largely unknown. Here we used in vivo two-photon microscopy in awake head-fixed mice to assess the role of spontaneous vasomotion in paravascular clearance. Vasomotion correlated with paravascular clearance of fluorescent dextran from the interstitial fluid. Increasing the amplitude of vasomotion by means of visually evoked vascular responses resulted in increased clearance rates in the visual cortex of awake mice. Evoked vascular reactivity was impaired in mice with CAA, which corresponded to slower clearance rates. Our findings suggest that low-frequency arteriolar oscillations drive drainage of solutes. Targeting naturally occurring vasomotion in patients with CAA or AD may be a promising early therapeutic option for prevention of Aβ accumulation in the brain.
Collapse
Affiliation(s)
- Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA.
| | - Steven S Hou
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA
| | - Maria Calvo-Rodriguez
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA
| | - Michal Arbel-Ornath
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA
| | - Austin C Snyder
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA
| | - Matthew P Frosch
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA; Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Brian J Bacskai
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA 02129, USA
| |
Collapse
|
49
|
From Stroke to Dementia: a Comprehensive Review Exposing Tight Interactions Between Stroke and Amyloid-β Formation. Transl Stroke Res 2019; 11:601-614. [PMID: 31776837 PMCID: PMC7340665 DOI: 10.1007/s12975-019-00755-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 01/13/2023]
Abstract
Stroke and Alzheimer’s disease (AD) are cerebral pathologies with high socioeconomic impact that can occur together and mutually interact. Vascular factors predisposing to cerebrovascular disease have also been specifically associated with development of AD, and acute stroke is known to increase the risk to develop dementia. Despite the apparent association, it remains unknown how acute cerebrovascular disease and development of AD are precisely linked and act on each other. It has been suggested that this interaction is strongly related to vascular deposition of amyloid-β (Aβ), i.e., cerebral amyloid angiopathy (CAA). Furthermore, the blood–brain barrier (BBB), perivascular space, and the glymphatic system, the latter proposedly responsible for the drainage of solutes from the brain parenchyma, may represent key pathophysiological pathways linking stroke, Aβ deposition, and dementia. In this review, we propose a hypothetic connection between CAA, stroke, perivascular space integrity, and dementia. Based on relevant pre-clinical research and a few clinical case reports, we speculate that impaired perivascular space integrity, inflammation, hypoxia, and BBB breakdown after stroke can lead to accelerated deposition of Aβ within brain parenchyma and cerebral vessel walls or exacerbation of CAA. The deposition of Aβ in the parenchyma would then be the initiating event leading to synaptic dysfunction, inducing cognitive decline and dementia. Maintaining the clearance of Aβ after stroke could offer a new therapeutic approach to prevent post-stroke cognitive impairment and development into dementia.
Collapse
|
50
|
Lendahl U, Nilsson P, Betsholtz C. Emerging links between cerebrovascular and neurodegenerative diseases-a special role for pericytes. EMBO Rep 2019; 20:e48070. [PMID: 31617312 PMCID: PMC6831996 DOI: 10.15252/embr.201948070] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative and cerebrovascular diseases cause considerable human suffering, and therapy options for these two disease categories are limited or non-existing. It is an emerging notion that neurodegenerative and cerebrovascular diseases are linked in several ways, and in this review, we discuss the current status regarding vascular dysregulation in neurodegenerative disease, and conversely, how cerebrovascular diseases are associated with central nervous system (CNS) degeneration and dysfunction. The emerging links between neurodegenerative and cerebrovascular diseases are reviewed with a particular focus on pericytes-important cells that ensheath the endothelium in the microvasculature and which are pivotal for blood-brain barrier function and cerebral blood flow. Finally, we address how novel molecular and cellular insights into pericytes and other vascular cell types may open new avenues for diagnosis and therapy development for these important diseases.
Collapse
Affiliation(s)
- Urban Lendahl
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
- Department of Neurobiology, Care Sciences and SocietyDivision of NeurogeriatricsCenter for Alzheimer ResearchKarolinska InstitutetSolnaSweden
- Integrated Cardio Metabolic Centre (ICMC)HuddingeSweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and SocietyDivision of NeurogeriatricsCenter for Alzheimer ResearchKarolinska InstitutetSolnaSweden
| | - Christer Betsholtz
- Integrated Cardio Metabolic Centre (ICMC)HuddingeSweden
- Department of Immunology, Genetics and PathologyRudbeck LaboratoryUppsala UniversityUppsalaSweden
- Department of MedicineKarolinska InstitutetHuddingeSweden
| |
Collapse
|