1
|
Adams JA, Uryash A, Lopez JR. Harnessing Passive Pulsatile Shear Stress for Alzheimer's Disease Prevention and Intervention. J Alzheimers Dis 2024; 98:387-401. [PMID: 38393906 DOI: 10.3233/jad-231010] [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] [Indexed: 02/25/2024]
Abstract
Alzheimer's disease (AD) affects more than 40 million people worldwide and is the leading cause of dementia. This disease is a challenge for both patients and caregivers and puts a significant strain on the global healthcare system. To address this issue, the Lancet Commission recommends focusing on reducing modifiable lifestyle risk factors such as hypertension, diabetes, and physical inactivity. Passive pulsatile shear stress (PPSS) interventions, which use devices like whole-body periodic acceleration, periodic acceleration along the Z-axis (pGz), and the Jogging Device, have shown significant systemic and cellular effects in preclinical and clinical models which address these modifiable risks factors. Based on this, we propose that PPSS could be a potential non-pharmacological and non-invasive preventive or therapeutic strategy for AD. We perform a comprehensive review of the biological basis based on all publications of PPSS using these devices and demonstrate their effects on the various aspects of AD. We draw from this comprehensive analysis to support our hypothesis. We then delve into the possible application of PPSS as an innovative intervention. We discuss how PPSS holds promise in ameliorating hypertension and diabetes while mitigating physical inactivity, potentially offering a holistic approach to AD prevention and management.
Collapse
Affiliation(s)
- Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
2
|
Mamelak M. The Alzheimer's Disease Brain, Its Microvasculature, and NADPH Oxidase. J Alzheimers Dis 2024; 99:S109-S118. [PMID: 37599534 DOI: 10.3233/jad-230415] [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] [Indexed: 08/22/2023]
Abstract
The deterioration of the brain's microvasculature, particularly in the hippocampus, appears to be a very early event in the development of Alzheimer's disease (AD), preceding even the deposition of amyloid-β. A damaged microvasculature reduces the supply of oxygen and glucose to this region and limits the production of energy, ATP. The damage may be a function of the rise with age in the expression and activity of NADPH oxidase (NOX) in these microvessels. This rise renders these vessels vulnerable to the effects of oxidative stress and inflammation. The rise in NOX activity with age is even more marked in the AD brain where an inverse correlation has been demonstrated between NOX activity and cognitive ability. Apocynin, a putative NOX inhibitor, has been shown to block the damaging effects of NOX activation. Apocynin acts as a strong scavenger of H2O2, and as a weak scavenger of superoxide. Like apocynin, sodium oxybate (SO) has also been shown to block the toxic effects of NOX activation. The application of SO generates NADPH and ATP. SO inhibits oxidative stress and maintains normal cerebral ATP levels under hypoxic conditions. Moreover, it acts epigenetically to attenuate the expression of NOX. SO may delay the onset and slow the progress of AD by suppling energy and maintaining an antioxidative environment in the brain throughout the night. The slow wave activity produced by SO may also activate the glymphatic system and promote the clearance of amyloid-β from the brain.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Maccallini C, Amoroso R. Neuronal Nitric Oxide Synthase and Post-Translational Modifications in the Development of Central Nervous System Diseases: Implications and Regulation. Molecules 2023; 28:6691. [PMID: 37764469 PMCID: PMC10538099 DOI: 10.3390/molecules28186691] [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: 08/26/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
In the Central Nervous System (CNS), Nitric Oxide (NO) is mainly biosynthesized by neuronal Nitric Oxide Synthase (nNOS). The dysregulated activation of nNOS in neurons is critical in the development of different conditions affecting the CNS. The excessive production of NO by nNOS is responsible for a number of proteins' post-translational modifications (PTMs), which can lead to aberrant biochemical pathways, impairing CNS functions. In this review, we briefly revise the main implications of dysregulated nNOS in the progression of the most prevalent CNS neurodegenerative disorders, i.e., Alzheimer's disease (AD) and Parkinson's disease, as well as in the development of neuronal disorders. Moreover, a specific focus on compounds able to modulate nNOS activity as promising therapeutics to tackle different neuronal diseases is presented.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | | |
Collapse
|
4
|
Shields KL, Jarrett CL, Bisconti AV, Park SH, Craig JC, Broxterman RM, Richardson RS. Preserved endothelium-independent vascular function with aging in men and women: evidence from the peripheral and cerebral vasculature. J Appl Physiol (1985) 2023; 135:559-571. [PMID: 37391885 PMCID: PMC10538978 DOI: 10.1152/japplphysiol.00571.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023] Open
Abstract
In the peripheral and cerebral vasculature, the impact of aging and sex on the endothelial-independent functional capacity of vascular smooth muscle cells (VSMCs) is not well understood, nor is it known whether such VSMC functions in these vascular beds reflect one another. Therefore, endothelium-independent dilation, at both the conduit (Δ diameter) and microvascular (Δ vascular conductance, VC) level, elicited by sublingual nitroglycerin (NTG, 0.8 mg of Nitrostat), compared with sham-delivery (control), was assessed using Doppler ultrasound in the popliteal (PA) and middle cerebral (MCA) artery of 20 young [23 ± 4 yr, 10 males (YM)/10 females (YF)] and 21 old [69 ± 5 yr, 11 males (OM)/10 females (OF)] relatively healthy adults. In the PA, compared with zero, NTG significantly increased diameter in all groups (YM: 0.29 ± 0.13, YF: 0.35 ± 0.26, OM: 0.30 ± 0.18, OF: 0.31 ± 0.14 mm), while control did not. The increase in VC only achieved significance in the OF (0.22 ± 0.31 mL/min/mmHg). In the MCA, compared with zero, NTG significantly increased diameter and VC in all groups (YM: 0.89 ± 0.30, 1.06 ± 1.28; YF: 0.97 ± 0.31, 1.84 ± 1.07; OM: 0.90 ± 0.42, 0.72 ± 0.99; OF: 0.74 ± 0.32, 1.19 ± 1.18, mm and mL/min/mmHg, respectively), while control did not. There were no age or sex differences or age-by-sex interactions for both the NTG-induced PA and MCA dilation and VC. In addition, PA and MCA dilation and VC responses to NTG were not related when grouped by age, sex, or as all subjects (r = 0.04-0.44, P > 0.05). Thus, peripheral and cerebral endothelial-independent VSMC function appears to be unaffected by age or sex, and variations in such VSMC function in one of these vascular beds are not reflected in the other.NEW & NOTEWORTHY To confidently explain peripheral and cerebral vascular dysfunction, it is essential to have a clear understanding of the endothelial-independent function of VSMCs across age and sex. By assessing endothelium-independent dilation using sublingual nitroglycerin, endothelial-independent VSMC function in the periphery (popliteal artery), and in the cerebral circulation (middle cerebral artery), was not different due to age or sex. In addition, endothelial-independent VSMC function in one of these vascular beds is not reflected in the other.
Collapse
Affiliation(s)
- Katherine L Shields
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Catherine L Jarrett
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Angela V Bisconti
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Soung Hun Park
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Jesse C Craig
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Ryan M Broxterman
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Center on Aging, University of Utah, Salt Lake City, Utah, United States
| | - Russell S Richardson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Center on Aging, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
5
|
Gullotta GS, Costantino G, Sortino MA, Spampinato SF. Microglia and the Blood-Brain Barrier: An External Player in Acute and Chronic Neuroinflammatory Conditions. Int J Mol Sci 2023; 24:ijms24119144. [PMID: 37298096 DOI: 10.3390/ijms24119144] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system that guarantee immune surveillance and exert also a modulating role on neuronal synaptic development and function. Upon injury, microglia get activated and modify their morphology acquiring an ameboid phenotype and pro- or anti-inflammatory features. The active role of microglia in blood-brain barrier (BBB) function and their interaction with different cellular components of the BBB-endothelial cells, astrocytes and pericytes-are described. Here, we report the specific crosstalk of microglia with all the BBB cell types focusing in particular on the involvement of microglia in the modulation of BBB function in neuroinflammatory conditions that occur in conjunction with an acute event, such as a stroke, or in a slow neurodegenerative disease, such as Alzheimer's disease. The potential of microglia to exert a dual role, either protective or detrimental, depending on disease stages and environmental conditioning factors is also discussed.
Collapse
Affiliation(s)
- Giorgia Serena Gullotta
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuseppe Costantino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Ph.D. Program in Neuroscience and Education, DISTUM, University of Foggia, 71121 Foggia, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | | |
Collapse
|
6
|
Abstract
eNOS (endothelial nitric oxide synthase) is critically important enzyme responsible for regulation of cardiovascular homeostasis. Under physiological conditions, constitutive eNOS activity and production of endothelial nitric oxide (NO) exert essential neurovascular protective functions. In this review, we first discuss the roles of endothelial NO in prevention of neuronal amyloid accumulation and formation of neurofibrillary tangles, hallmarks of Alzheimer disease pathology. Next, we review existing evidence suggesting that NO released from endothelium prevents activation of microglia, stimulates glycolysis in astrocytes, and increases biogenesis of mitochondria. We also address major risk factors for cognitive impairment including aging and ApoE4 (apolipoprotein 4) genotype with focus on their detrimental effects on eNOS/NO signaling. Relevant to this review, recent studies suggested that aged eNOS heterozygous mice are unique model of spontaneous cerebral small vessel disease. In this regard, we review contribution of dysfunctional eNOS to deposition of Aβ (amyloid-β) into blood vessel wall leading to development of cerebral amyloid angiopathy. We conclude that endothelial dysfunction manifested by the loss of neurovascular protective functions of NO may significantly contribute to development of cognitive impairment.
Collapse
Affiliation(s)
- Zvonimir S. Katusic
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Livius V. d’Uscio
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Tongrong He
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
7
|
Takami Y, Wang C, Nakagami H, Yamamoto K, Nozato Y, Imaizumi Y, Nagasawa M, Takeshita H, Nakajima T, Takeda S, Takeya Y, Kaneda Y, Rakugi H. Novel pathophysiological roles of α-synuclein in age-related vascular endothelial dysfunction. FASEB J 2022; 36:e22555. [PMID: 36125010 DOI: 10.1096/fj.202101621r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 11/11/2022]
Abstract
Although α-synuclein (SNCA) is a well-known pathological molecule involved in synucleinopathy in neurons, its physiological roles remain largely unknown. We reported that serum SNCA levels have a close inverse correlation with blood pressure and age, which indicates the involvement of SNCA in age-related endothelial dysfunction. Therefore, this study aimed to elucidate the molecular functions of SNCA in the endothelium. We confirmed that SNCA was expressed in and secreted from endothelial cells (ECs). Exogenous treatment with recombinant SNCA (rSNCA) activated the Akt-eNOS axis and increased nitric oxide production in ECs. Treatment with rSNCA also suppressed TNF-α- and palmitic acid-induced NF-κB activation, leading to the suppression of VCAM-1 upregulation and restoration of eNOS downregulation in ECs. As for endogenous SNCA expression, replicative senescence resulted in the attenuation of SNCA expression in cultured ECs, similar to the effects of physiological aging on mice aortas. The siRNA-mediated silencing of SNCA consistently resulted in senescent phenotypes, such as eNOS downregulation, increased β-gal activity, decreased Sirt1 expression, and increased p53 expression, in ECs. Ex vivo assessment of endothelial functions using aortic rings revealed impaired endothelium-dependent acetylcholine-induced relaxation in SNCA knockout (KO) mice. Furthermore, SNCA KO mice, especially those on a high-fat diet, displayed elevated blood pressure compared with wild-type mice; this could be eNOS dysfunction-dependent because of the lower difference caused by L-NAME administration. These results indicate that exogenous and endogenous SNCA in ECs might physiologically maintain vascular integrity, and age-related endothelial dysfunction might be partially ascribed to loss-of-function of SNCA in ECs.
Collapse
Affiliation(s)
- Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Cheng Wang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Imaizumi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motonori Nagasawa
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hikari Takeshita
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsuneo Nakajima
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka, Japan.,Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Yasushi Takeya
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
8
|
Adams JA, Lopez JR, Nadkarni V, Zolkipli‐Cunningham Z, Ischiropoulos H, Sackner MA. The effects of a motorized passive simulated jogging device on descent of the arterial pulse waveform dicrotic notch: A single arm placebo-controlled cross-over trial. Physiol Rep 2022; 10:e15418. [PMID: 35924333 PMCID: PMC9350470 DOI: 10.14814/phy2.15418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 11/06/2023] Open
Abstract
Whole Body Periodic Acceleration (WBPA, pGz), is a bed that moves the body headward to forward, adds pulses to the circulation inducing descent of the dicrotic notch (DN) on the pulse waveform with an increase in a/b ratio (a = the height of the pulse waveform and b = the height of the secondary wave). Since the WBPA is large, heavy, and non-portable, we engineered a portable device (Jogging Device, JD). JD simulates passive jogging and introduces pulsations to the circulation. We hypothesized that JD would increase the a/b ratio during and after its use. In Study A, a single-arm placebo-controlled cross-over trial was conducted in24 adults (53.8 ± 14.4 years) using JD or control (CONT) for 30 min. Blood pressure (BPs and BPd) and photoplethysmograph pulse (a/b) were measured at baseline (BL), during 30 min of JD or CONT, and 5 and 60 min after. In Study B (n = 20, 52.2 ± 7 years), a single-arm observational trial of 7 consecutive days of JD on BP and a/b, measured at BL, and after 7 days of JD and 48 and 72 hr after its discontinuation. In Study A, BPs, and BPd decreased during JD by 13% and 16%, respectively, while in CONT both increased by 2% and 2.5%, respectively. The a/b increased by 2-fold and remained greater than 2-fold at all-time points, with no change in a/b during CONT. In Study B, BPs and BPd decreased by 9% and remained below BL, at 72 hr after discontinuation of JD. DN descent also occurred after 7 days of JD with a/b increase of 80% and remained elevated by 60% for at least 72 h. JD improves acute and longer-term vascular hemodynamics with an increase in a/b, consistent with increased effects of nitric oxide (NO). JD may have significant clinical and public health implications.
Collapse
Affiliation(s)
- Jose A. Adams
- Division NeonatologyMt Sinai Medical Center of Greater MiamiMiami BeachFloridaUSA
| | - Jose R. Lopez
- Department of ResearchMt Sinai Medical Center of Greater MiamiMiami BeachFloridaUSA
| | - Vinay Nadkarni
- Anesthesiology, Critical Care, and Pediatrics, The Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Zarazuela Zolkipli‐Cunningham
- Mitochondrial Medicine Frontier Program (MMFP), Center for Mitochondrial and Epigenomic Medicine (CMEM), Division of Human Genetics, The Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute and Division of Neonatology, Departments of Pediatrics and Systems Pharmacology and Translational Therapeutics, the Raymond and Ruth Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Marvin A. Sackner
- Department of ResearchMt Sinai Medical Center of Greater MiamiMiami BeachFloridaUSA
| |
Collapse
|
9
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
10
|
Angermann S, Günthner R, Hanssen H, Lorenz G, Braunisch MC, Steubl D, Matschkal J, Kemmner S, Hausinger R, Block Z, Haller B, Heemann U, Kotliar K, Grimmer T, Schmaderer C. Cognitive impairment and microvascular function in end-stage renal disease. Int J Methods Psychiatr Res 2022; 31:e1909. [PMID: 35290686 PMCID: PMC9159686 DOI: 10.1002/mpr.1909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Hemodialysis patients show an approximately threefold higher prevalence of cognitive impairment compared to the age-matched general population. Impaired microcirculatory function is one of the assumed causes. Dynamic retinal vessel analysis is a quantitative method for measuring neurovascular coupling and microvascular endothelial function. We hypothesize that cognitive impairment is associated with altered microcirculation of retinal vessels. METHODS 152 chronic hemodialysis patients underwent cognitive testing using the Montreal Cognitive Assessment. Retinal microcirculation was assessed by Dynamic Retinal Vessel Analysis, which carries out an examination recording retinal vessels' reaction to a flicker light stimulus under standardized conditions. RESULTS In unadjusted as well as in adjusted linear regression analyses a significant association between the visuospatial executive function domain score of the Montreal Cognitive Assessment and the maximum arteriolar dilation as response of retinal arterioles to the flicker light stimulation was obtained. CONCLUSION This is the first study determining retinal microvascular function as surrogate for cerebral microvascular function and cognition in hemodialysis patients. The relationship between impairment in executive function and reduced arteriolar reaction to flicker light stimulation supports the involvement of cerebral small vessel disease as contributing factor for the development of cognitive impairment in this patient population and might be a target for noninvasive disease monitoring and therapeutic intervention.
Collapse
Affiliation(s)
- Susanne Angermann
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Roman Günthner
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Henner Hanssen
- Department of Sport, Exercise and HealthUniversity of BaselBaselSwitzerland
| | - Georg Lorenz
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Matthias C. Braunisch
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Dominik Steubl
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Julia Matschkal
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Stephan Kemmner
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
- Transplant CenterUniversity Hospital MunichLudwig‐Maximilians‐University (LMU)MunichGermany
| | - Renate Hausinger
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Zenonas Block
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and EpidemiologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Uwe Heemann
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| | - Konstantin Kotliar
- Department of Medical Engineering and TechnomathematicsFH Aachen University of Applied SciencesCampus JülichJülichGermany
| | - Timo Grimmer
- Department of Psychiatry and PsychotherapyKlinikum rechts der IsarTechnische Universität MünchenMunichGermany
| | - Christoph Schmaderer
- Department of NephrologyTechnical University of MunichSchool of Medicine, Klinikum rechts der IsarMunchenGermany
| |
Collapse
|
11
|
Alghadir AH, Gabr SA, Almomani M, Almomani F, Tse C. Adiponectin and Nitric Oxide Deficiency-Induced Cognitive Impairment in Fatigued Home-Resident in Mature and Older Adults: A Case-Control Study. Pain Res Manag 2022; 2022:7480579. [PMID: 35600795 PMCID: PMC9117056 DOI: 10.1155/2022/7480579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 11/29/2021] [Accepted: 04/19/2022] [Indexed: 11/18/2022]
Abstract
Objective The present study explores the underlying factors of cognitive abilities in relation to the expression of adiponectin and nitric oxide, fatigue, and other cofounder variables such as physical activity, diabetes, and adiposity status in healthy home-resident mature and older adults. Background Fatigue has been shown to be correlated with many metabolic and psychiatric conditions, such as cognitive, neurological, musculoskeletal, and hormonal disorders, as well as physical and unhealthy lifestyles. Methods A total of 85 home residents aged 50-85 years participated in this case-control study. Mental, fatigue, and pain status were assessed by the cognitive assessment (LOTCA), fatigue questionnaire (CIS20r), and pain score (0-10). VO2 max and the prevalidated global physical activity questionnaire were used to estimate physical status. The levels of adiponectin, nitric oxide (NO), and variables related to diabetes, such as blood sugar and glycated hemoglobin (HbA1c %), were assessed using ELISA and spectrophotometric immunoassays. Results The participants were classified according to the CIS-fatigue score into two groups: the healthy group (n = 40) and the fatigue group (n = 45). In fatigued subjects, LOTCA scores as a measure of cognitive performance significantly decreased (65.97 ± 7.17; P = 0.01) as compared with healthy subjects (LOTCA scores, 94.2 ± 7.5). The results of cognitive performance domains (LOTCA seven-subset scores) showed a significant decrease in the scores of orientation, visual perception, spatial perception, motor praxis, vasomotor organization, thinking operations, attention, and concentration in older subjects with fatigue compared with healthy subjects. In addition, pain scores significantly increased, and the expression of both nitric oxide (NO) and adiponectin significantly reduced in older adults with fatigue as compared with healthy controls. The decline in cognitive abilities among older adults with fatigue is significantly associated with the CIS-fatigue score, sedentary lifestyle, obesity, pain status, diabetes, and reduction in the levels of nitric oxide (NO), and adiponectin. Moreover, in fatigued cases, the expression of both NO and adiponectin was significantly correlated with CIS-fatigue score, physical activity, obesity, and diabetes, which indicates its availability as diagnostic markers for cognition in mature and older adults with fatigue. Conclusion In the present study, the data concluded that cognitive abilities were significantly associated with the lower expression of adiponectin and NO as endothelial vascular markers in association with fatigue among home-resident older adults. In addition, the reduction in cognition was significantly affected by other parameters, such as diabetes, obesity, and unhealthy sedentary life activities. Moreover, the results might recommend the use of cellular adiponectin and NO as diagnostic indicators of cognitive abilities in fatigued mature and older adults. However, more studies on larger sample sizes are required.
Collapse
Affiliation(s)
- Ahmad H. Alghadir
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Sami A. Gabr
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Murad Almomani
- ORL-HNS Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fidaa Almomani
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Cynthia Tse
- Faculty of Movement and Rehabilitation Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Hernando S, Nikolakopoulou P, Voulgaris D, Hernandez RM, Igartua M, Herland A. Dual effect of TAT functionalized DHAH lipid nanoparticles with neurotrophic factors in human BBB and microglia cultures. Fluids Barriers CNS 2022; 19:22. [PMID: 35300705 PMCID: PMC8928663 DOI: 10.1186/s12987-022-00315-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023] Open
Abstract
Background Neurodegenerative diseases (NDs) are an accelerating global health problem. Nevertheless, the stronghold of the brain- the blood–brain barrier (BBB) prevents drug penetrance and dwindles effective treatments. Therefore, it is crucial to identify Trojan horse-like drug carriers that can effectively cross the blood–brain barrier and reach the brain tissue. We have previously developed polyunsaturated fatty acids (PUFA)-based nanostructured lipid carriers (NLC), namely DHAH-NLC. These carriers are modulated with BBB-permeating compounds such as chitosan (CS) and trans-activating transcriptional activator (TAT) from HIV-1 that can entrap neurotrophic factors (NTF) serving as nanocarriers for NDs treatment. Moreover, microglia are suggested as a key causative factor of the undergoing neuroinflammation of NDs. In this work, we used in vitro models to investigate whether DHAH-NLCs can enter the brain via the BBB and investigate the therapeutic effect of NTF-containing DHAH-NLC and DHAH-NLC itself on lipopolysaccharide-challenged microglia. Methods We employed human induced pluripotent stem cell-derived brain microvascular endothelial cells (BMECs) to capitalize on the in vivo-like TEER of this BBB model and quantitatively assessed the permeability of DHAH-NLCs. We also used the HMC3 microglia cell line to assess the therapeutic effect of NTF-containing DHAH-NLC upon LPS challenge. Results TAT-functionalized DHAH-NLCs successfully crossed the in vitro BBB model, which exhibited high transendothelial electrical resistance (TEER) values (≈3000 Ω*cm2). Specifically, the TAT-functionalized DHAH-NLCs showed a permeability of up to 0.4% of the dose. Furthermore, using human microglia (HMC3), we demonstrate that DHAH-NLCs successfully counteracted the inflammatory response in our cultures after LPS challenge. Moreover, the encapsulation of glial cell-derived neurotrophic factor (GNDF)-containing DHAH-NLCs (DHAH-NLC-GNDF) activated the Nrf2/HO-1 pathway, suggesting the triggering of the endogenous anti-oxidative system present in microglia. Conclusions Overall, this work shows that the TAT-functionalized DHAH-NLCs can cross the BBB, modulate immune responses, and serve as cargo carriers for growth factors; thus, constituting an attractive and promising novel drug delivery approach for the transport of therapeutics through the BBB into the brain. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00315-1.
Collapse
Affiliation(s)
- Sara Hernando
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institutet and KTH Royal Institute of Technology, 171 77, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.,NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029, Madrid, Spain.,Bioaraba, NanoBioCel Research Group, 01006, Vitoria-Gasteiz, Spain
| | - Polyxeni Nikolakopoulou
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institutet and KTH Royal Institute of Technology, 171 77, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Dimitrios Voulgaris
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institutet and KTH Royal Institute of Technology, 171 77, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.,Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.,Division of Micro and Nanosystems, KTH Royal Institute of Technology, 171 77, Stockholm, Sweden
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029, Madrid, Spain.,Bioaraba, NanoBioCel Research Group, 01006, Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain. .,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029, Madrid, Spain. .,Bioaraba, NanoBioCel Research Group, 01006, Vitoria-Gasteiz, Spain.
| | - Anna Herland
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institutet and KTH Royal Institute of Technology, 171 77, Stockholm, Sweden. .,Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden. .,Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.
| |
Collapse
|
13
|
An activatable fluorescent probe for imaging endogenous nitric oxide via the eNOS enzymatic pathway. Bioorg Med Chem Lett 2022; 59:128544. [DOI: 10.1016/j.bmcl.2022.128544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 11/22/2022]
|
14
|
Yuan M, Wang Y, Wang S, Huang Z, Jin F, Zou Q, Li J, Pu Y, Cai Z. Bioenergetic Impairment in the Neuro-Glia-Vascular Unit: An Emerging Physiopathology during Aging. Aging Dis 2021; 12:2080-2095. [PMID: 34881087 PMCID: PMC8612602 DOI: 10.14336/ad.2021.04017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
An emerging concept termed the "neuro-glia-vascular unit" (NGVU) has been established in recent years to understand the complicated mechanism of multicellular interactions among vascular cells, glial cells, and neurons. It has been proverbially reported that the NGVU is significantly associated with neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Physiological aging is an inevitable progression associated with oxidative damage, bioenergetic alterations, mitochondrial dysfunction, and neuroinflammation, which is partially similar to the pathology of AD. Thus, senescence is regarded as the background for the development of neurodegenerative diseases. With the exacerbation of global aging, senescence is an increasingly serious problem in the medical field. In this review, the coupling of each component, including neurons, glial cells, and vascular cells, in the NGVU is described in detail. Then, various mechanisms of age-dependent impairment in each part of the NGVU are discussed. Moreover, the potential bioenergetic alterations between different cell types in the NGVU are highlighted, which seems to be an emerging physiopathology associated with the aged brain. Bioenergetic intervention in the NGVU may be a new direction for studies on delaying or diminishing aging in the future.
Collapse
Affiliation(s)
- Minghao Yuan
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Yangyang Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Shengyuan Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Zhenting Huang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Feng Jin
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Qian Zou
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Jing Li
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Yinshuang Pu
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Zhiyou Cai
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
István L, Czakó C, Élő Á, Mihály Z, Sótonyi P, Varga A, Ungvári Z, Csiszár A, Yabluchanskiy A, Conley S, Csipő T, Lipecz Á, Kovács I, Nagy ZZ. Imaging retinal microvascular manifestations of carotid artery disease in older adults: from diagnosis of ocular complications to understanding microvascular contributions to cognitive impairment. GeroScience 2021; 43:1703-1723. [PMID: 34100219 PMCID: PMC8492863 DOI: 10.1007/s11357-021-00392-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Carotid artery stenosis (CAS) is a consequence of systemic atherosclerotic disease affecting the aging populations of the Western world. CAS is frequently associated with cognitive impairment. However, the mechanisms contributing to the development of vascular cognitive impairment (VCI) associated with CAS are multifaceted and not fully understood. In addition to embolization and decreased blood flow due to the atherosclerotic lesion in the carotid artery, microcirculatory dysfunction in the cerebral circulation also plays a critical role in CAS-related VCI. To better understand the microvascular contributions to cognitive decline associated with CAS and evaluate microvascular protective effects of therapeutic interventions, it is essential to examine the structural and functional changes of the microvessels in the central nervous system (CNS). However, there are some limitations of in vivo brain vascular imaging modalities. The retinal microvasculature provides a unique opportunity to study pathogenesis of cerebral small vessel disease and VCI, because the cerebral circulation and the retinal circulation share similar anatomy, physiology and embryology. Similar microvascular pathologies may manifest in the brain and the retina, thus ocular examination can be used as a noninvasive screening tool to investigate pathological changes in the CNS associated with CAS. In this review, ocular signs of CAS and the retinal manifestations of CAS-associated microvascular dysfunction are discussed. The advantages and limitation of methods that are capable of imaging the ocular circulation (including funduscopy, fluorescein angiography, Doppler sonography, optical coherence tomography [OCT] and optical coherence tomography angiography [OCTA]) are discussed. The potential use of dynamic retinal vessel analysis (DVA), which allows for direct visualization of neurovascular coupling responses in the CNS, for understanding microvascular contributions to cognitive decline in CAS patients is also considered.
Collapse
Affiliation(s)
- Lilla István
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Cecilia Czakó
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Ágnes Élő
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Zsuzsanna Mihály
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Péter Sótonyi
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Andrea Varga
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Zoltán Ungvári
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Anna Csiszár
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamás Csipő
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ágnes Lipecz
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Josa Andras Hospital, Nyiregyhaza, Hungary
| | - Illés Kovács
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary.
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA.
- Department of Clinical Ophtalmology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary.
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| |
Collapse
|
16
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
17
|
Zhong M, Peng J, Xiang L, Yang X, Wang X, Zhu Y. Epigallocatechin Gallate (EGCG) Improves Anti-Angiogenic State, Cell Viability, and Hypoxia-Induced Endothelial Dysfunction by Downregulating High Mobility Group Box 1 (HMGB1) in Preeclampsia. Med Sci Monit 2020; 26:e926924. [PMID: 33056943 PMCID: PMC7574359 DOI: 10.12659/msm.926924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Preeclampsia (PE) is a serious complication of pregnancy with no effective therapy. This study assessed whether epigallocatechin gallate (EGCG) could reduce the production of anti-angiogenic factors, improve cell viability, and suppress endothelial dysfunction in vitro via regulating high mobility group box 1 (HMGB1) in preeclampsia. Material/Methods Human umbilical vein endothelial cells (HUVECs) grown in conditioned medium from hypoxic JEG-3 cells were used to investigate the effects of EGCG on anti-angiogenic state, cell viability, and markers of endothelial dysfunction. To confirm that EGCG exerted its effects via HMGB1, we also examined the impact of EGCG on anti-angiogenic state, cell viability, and endothelial dysfunction following HMGB1 treatment in vitro. Results EGCG inhibited HMGB1 expression in hypoxic trophoblast cells in a dose-dependent manner. In addition, EGCG relieved anti-angiogenic state and endothelial dysfunction in hypoxic trophoblast cells by downregulating HMGB1. Moreover, EGCG dose-dependently promoted cell proliferation by downregulating HMGB1. Conclusions Taken together, our data show the protective role of EGCG in preeclampsia and revealed EGCG-mediated effects on the production of anti-angiogenic factors, cell viability, and endothelial dysfunction through downregulating HMGB1. These observations suggest that EGCG is a novel therapeutic candidate for preeclampsia.
Collapse
Affiliation(s)
- Min Zhong
- Department of Obstetrics, People's Hospital of Longhua Shenzhen, Shenzhen, Guangdong, China (mainland)
| | - Julan Peng
- Department of Obstetrics, People's Hospital of Longhua Shenzhen, Shenzhen, Guangdong, China (mainland)
| | - Lanhua Xiang
- Department of Obstetrics, People's Hospital of Longhua Shenzhen, Shenzhen, Guangdong, China (mainland)
| | - Xinhuang Yang
- Department of Obstetrics, People's Hospital of Longhua Shenzhen, Shenzhen, Guangdong, China (mainland)
| | - Xianghua Wang
- Department of Obstetrics, People's Hospital of Longhua Shenzhen, Shenzhen, Guangdong, China (mainland)
| | - Yanbin Zhu
- Department of Obstetrics, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China (mainland)
| |
Collapse
|
18
|
Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, Hughes TM, Daemen MJ, Williamson JD, González HM, Schneider J, Wellington CL, Katusic ZS, Stoeckel L, Koenig JI, Corriveau RA, Fine L, Galis ZS, Reis J, Wright JD, Chen J. Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement 2020; 16:1714-1733. [PMID: 33030307 DOI: 10.1002/alz.12157] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are characterized by the aging neurovascular unit being confronted with and failing to cope with biological insults due to systemic and cerebral vascular disease, proteinopathy including Alzheimer's biology, metabolic disease, or immune response, resulting in cognitive decline. This report summarizes the discussion and recommendations from a working group convened by the National Heart, Lung, and Blood Institute and the National Institute of Neurological Disorders and Stroke to evaluate the state of the field in VCID research, identify research priorities, and foster collaborations. As discussed in this report, advances in understanding the biological mechanisms of VCID across the wide spectrum of pathologies, chronic systemic comorbidities, and other risk factors may lead to potential prevention and new treatment strategies to decrease the burden of dementia. Better understanding of the social determinants of health that affect risks for both vascular disease and VCID could provide insight into strategies to reduce racial and ethnic disparities in VCID.
Collapse
Affiliation(s)
| | | | | | - Sudha Seshadri
- University of Texas Health Science Center, San Antonio and Boston University, San Antonio, Texas, USA
| | - Ann McKee
- VA Boston Healthcare System and Boston University, Boston, Massachusetts, USA
| | | | - Steven M Greenberg
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristine Yaffe
- University of California, San Francisco, San Francisco, California, USA
| | | | - Chun Yuan
- University of Washington, Seattle, Washington, USA
| | - Timothy M Hughes
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mat J Daemen
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Luke Stoeckel
- National Institute on Aging, Bethesda, Maryland, USA
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Roderick A Corriveau
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Lawrence Fine
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Zorina S Galis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jared Reis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | | | - Jue Chen
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Hong J, Hong SG, Lee J, Park JY, Eriksen JL, Rooney BV, Park Y. Exercise training ameliorates cerebrovascular dysfunction in a murine model of Alzheimer's disease: role of the P2Y2 receptor and endoplasmic reticulum stress. Am J Physiol Heart Circ Physiol 2020; 318:H1559-H1569. [PMID: 32383993 DOI: 10.1152/ajpheart.00129.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cerebrovascular dysfunction is a critical risk factor for the pathogenesis of Alzheimer's disease (AD). The purinergic P2Y2 receptor and endoplasmic reticulum (ER) stress are tightly associated with vascular dysfunction and the pathogenesis of AD. However, the protective effects of exercise training on P2Y2 receptor- and ER stress-associated cerebrovascular dysfunction in AD are mostly unknown. Control (C57BL/6, CON) and AD (APP/PS1dE9, AD) mice underwent treadmill exercise training (EX). 2-MeS-ATP-induced dose-dependent vasoreactivity was determined by using a pressurized posterior cerebral artery (PCA) from 10-12-mo-old mice. Human brain microvascular endothelial cells (HBMECs) were exposed to laminar shear stress (LSS) at 20 dyn/cm2 for 30 min, 2 h, and 24 h. The expression of P2Y2 receptors, endothelial nitric oxide synthase (eNOS), and ER stress signaling were quantified by Western blot analysis. Notably, exercise converted ATP-induced vasoconstriction in the PCA from AD mice to vasodilation in AD+EX mice to a degree commensurate to the vascular reactivity observed in CON mice. Exercise reduced the expression of amyloid peptide precursor (APP) and increased the P2Y2 receptor and Akt/eNOS expression in AD mice brain. Mechanistically, LSS increased the expression of both P2Y2 receptor and eNOS protein in HBMECs, but these increases were blunted by a P2Y2 receptor antagonist in HBMECs. Exercise also reduced the expression of aberrant ER stress markers p-IRE1, p/t-eIF2α, and CHOP, as well as Bax/Bcl-2, in AD mice brain. Collectively, our results demonstrate for the first time that exercise mitigates cerebrovascular dysfunction in AD through modulating P2Y2 receptor- and ER stress-dependent endothelial dysfunction.NEW & NOTEWORTHY A limited study has investigated whether exercise training can improve cerebrovascular function in Alzheimer's disease. The novel findings of the study are that exercise training improves cerebrovascular dysfunction through enhancing P2Y2 receptor-mediated eNOS signaling and reducing ER stress-associated pathways in AD. These data suggest that exercise training, which regulates P2Y2 receptor and ER stress in AD brain, is a potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Junyoung Hong
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| | - Soon-Gook Hong
- Department of Kinesiology and Cardiovascular Research Center, Temple University, Philadelphia, Pennsylvania
| | - Jonghae Lee
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| | - Joon-Young Park
- Department of Kinesiology and Cardiovascular Research Center, Temple University, Philadelphia, Pennsylvania
| | - Jason L Eriksen
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas
| | - Bridgette V Rooney
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas.,Geocontrol Systems, Incorporated, Johnson Space Center, National Aeronautics and Space Administration, Houston, Texas
| | - Yoonjung Park
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| |
Collapse
|
20
|
D’Arrigo JS. Nanotargeting of Drug(s) for Delaying Dementia: Relevance of Covid-19 Impact on Dementia. Am J Alzheimers Dis Other Demen 2020; 35:1533317520976761. [PMID: 33307726 PMCID: PMC10623919 DOI: 10.1177/1533317520976761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
By incorporating appropriate drug(s) into lipid (biobased) nanocarriers, one obtains a combination therapeutic for dementia treatment that targets certain cell-surface scavenger receptors (mainly class B type I, or "SR-BI") and thereby crosses the blood-brain barrier. The cardiovascular risk factors for dementia trigger widespread inflammation -- which lead to neurodegeneration, gradual cognitive/memory decline, and eventually (late-onset) dementia. Accordingly, one useful strategy to delay dementia could be based upon nanotargeting drug(s), using lipid nanocarriers, toward a major receptor class responsible for inflammation-associated (cytokine-mediated) cell signaling events. At the same time, the immune response and excessive inflammation, commonly observed in the very recent human coronavirus (COVID-19) pandemic, may accelerate the progression of brain inflammatory neurodegeneration-which increases the probability of post-infection memory impairment and accelerating progression of Alzheimer's disease. Hence, the proposed multitasking combination therapeutic, using a (biobased) lipid nanocarrier, may also display greater effectiveness at different stages of dementia.
Collapse
Affiliation(s)
- Joseph S. D’Arrigo
- Cavitation-Control Technology Inc, Farmington, CT, USA. D’Arrigo is now with Cav-Con, Inc, Bellevue, WA, USA
| |
Collapse
|
21
|
Loiselle AR, Neustaeter A, de Kleine E, van Dijk P, Jansonius NM. Associations between tinnitus and glaucoma suggest a common mechanism: A clinical and population-based study. Hear Res 2019; 386:107862. [PMID: 31841861 DOI: 10.1016/j.heares.2019.107862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to determine if there is an association between tinnitus and glaucoma. We tested this by first completing a clinic-based cross-sectional questionnaire study in which we sent a series of tinnitus-related questions to glaucoma patients and healthy subjects, and then followed up with a large population-based cross-sectional study in which glaucoma and tinnitus were also assessed by questionnaire. For the clinical study, we received 209 responses from glaucoma patients and 109 responses from healthy subjects (primarily the spouses of the patients). For the population-based study, we evaluated 79,866 participants. Logistic regression models were used to test the relationship between glaucoma and tinnitus; the clinical study analysis was adjusted for age, gender, BMI, hypertension, and diabetes and the population-based study was adjusted for these same variables with the addition of socioeconomic status and subjective hearing loss. For the clinical study, glaucoma patients had an 85% increase in odds for tinnitus (adjusted OR 1.85, 95% CI 1.10 to 3.05). The effect did not depend on pretreatment intraocular pressure, and the associated symptoms were not pulsatile in nature. For the population-based study, glaucoma patients had a 19% increase in odds for tinnitus (adjusted OR 1.19, 95% CI 1.02 to 1.40). Overall, our results suggest that those with glaucoma are more likely to have tinnitus than those without glaucoma. These results provide hypotheses for a mechanism involved in both tinnitus and glaucoma. One possible mechanism could be vascular dysregulation due to impairment of nitric oxide production.
Collapse
Affiliation(s)
- Allison R Loiselle
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anna Neustaeter
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Emile de Kleine
- Department of Otorhinolaryngology/Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Pim van Dijk
- Department of Otorhinolaryngology/Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nomdo M Jansonius
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
22
|
Kraal AZ, Moll AC, Arvanitis NR, Ward KM, Dougherty RJ, Grove TB, Burghardt KJ, Ellingrod VL. Metabolic syndrome is negatively associated with cognition among endothelial nitric oxide synthase (eNOS)- 786C carriers in schizophrenia-spectrum disorders. J Psychiatr Res 2019; 117:142-147. [PMID: 31421598 PMCID: PMC6707862 DOI: 10.1016/j.jpsychires.2019.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/30/2019] [Accepted: 07/25/2019] [Indexed: 02/08/2023]
Abstract
Although metabolic syndrome and cognitive inefficiencies are well-described common complications of schizophrenia, their association remains inconsistent, potentially due to poorly understood mechanisms underlying their relationship. Variability in the endothelial nitric oxide synthase (eNOS) gene, specifically the T-786C variant, has been separately associated with cognition and metabolic syndrome, with worse outcomes for eNOS-786C carriers likely occurring via negative effects on blood vessel functioning. However, the interaction between eNOS and metabolic syndrome in cognition among adults with schizophrenia is unknown. This study aimed to test the main and interaction effects of the eNOS-786C allele in cognition using hierarchical regression analyses controlling for age, sex, education, race, and antipsychotic exposure. Metabolic syndrome, eNOS T-786C genotype, and cognitive performance were assessed in 226 community-dwelling participants with chronic schizophrenia-spectrum disorders. Results demonstrated a significant interaction between metabolic syndrome and the eNOS-786C allele. Specifically, among eNOS-786C carriers only, metabolic syndrome was independently associated with lower scores in processing speed and verbal fluency, and predicted 12.5% and 15.8% of variance in performance, respectively. These results suggest that the additive negative effects of eNOS-786C and metabolic syndrome on blood vessel functioning may be severe enough to negatively impact cognition. The finding that metabolic syndrome is associated with worse cognition only in the presence of the eNOS-786C allele may clarify extant inconsistencies in the literature. These findings provide preliminary evidence that may inform interventions to reduce cognitive morbidity among adults with schizophrenia.
Collapse
Affiliation(s)
- A. Zarina Kraal
- Department of Psychology, College of Literature, Science, and the Arts, University of Michigan,Department of Clinical Pharmacy, College of Pharmacy, University of Michigan
| | - Allison C. Moll
- Department of Psychology, College of Literature, Science, and the Arts, University of Michigan
| | - Nicole R. Arvanitis
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan
| | - Kristen M. Ward
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan
| | - Ryan J. Dougherty
- Department of Social Welfare, Luskin School of Public Affairs, University of California Los Angeles
| | - Tyler B. Grove
- Department of Psychiatry, School of Medicine, University of Michigan
| | - Kyle J. Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University
| | - Vicki L. Ellingrod
- Department of Psychology, College of Literature, Science, and the Arts, University of Michigan,Department of Clinical Pharmacy, College of Pharmacy, University of Michigan,Department of Psychiatry, School of Medicine, University of Michigan
| |
Collapse
|
23
|
Mizuno M, Aso K, Tsuzuki Y, Kitazawa T, Migita O, Hokuto I, Yamamoto H. A successful treatment of tadalafil in incontinentia pigmenti with pulmonary hypertension. Eur J Med Genet 2019; 63:103764. [PMID: 31518693 DOI: 10.1016/j.ejmg.2019.103764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 09/09/2019] [Indexed: 11/19/2022]
Abstract
We describe a female infant with incontinentia pigmenti complicated by severe pulmonary arterial hypertension that was markedly improved by tadalafil administration. The infant was referred to our institution because of neonatal seizures and generalized skin rash at the age of 1 day. She was diagnosed with incontinentia pigmenti on skin biopsy findings. In addition to incontinentia pigmenti, she had pulmonary arterial hypertension without structural heart disease. The pulmonary hypertension rapidly worsened at the age of 2 months and was confirmed by cardiac catheterization. The pulmonary artery pressure was equal to systemic pressure but it decreased in response to nitric oxide inhalation. We, therefore, initiated treatment with tadalafil of 1 mg/kg/day. The follow-up cardiac catheterization performed at 9 months revealed dramatic improvement in the pulmonary artery pressure. An IKBKG mutation with deletion of exons 4-10 was detected in the blood of both the patient and her mother. Our experience indicates that tadalafil may be beneficial in treating pulmonary arterial hypertension associated with incontinentia pigmenti.
Collapse
Affiliation(s)
- Masanori Mizuno
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Kentaro Aso
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshimitsu Tsuzuki
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomoko Kitazawa
- Department of Dermatology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ohsuke Migita
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Isamu Hokuto
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hitoshi Yamamoto
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
24
|
Bazzigaluppi P, Beckett TL, Koletar MM, Hill ME, Lai A, Trivedi A, Thomason L, Dorr A, Gallagher D, Librach CL, Joo IL, McLaurin J, Stefanovic B. Combinatorial Treatment Using Umbilical Cord Perivascular Cells and Aβ Clearance Rescues Vascular Function Following Transient Hypertension in a Rat Model of Alzheimer Disease. Hypertension 2019; 74:1041-1051. [PMID: 31476904 PMCID: PMC6739147 DOI: 10.1161/hypertensionaha.119.13187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is available in the text. Transient hypertension is a risk factor for Alzheimer disease (AD), but the effects of this interaction on brain vasculature are understudied. Addressing vascular pathology is a promising avenue to potentiate the efficacy of treatments for AD. We used arterial spin labeling magnetic resonance imaging to longitudinally assess brain vascular function and immunohistopathology to examine cerebrovascular remodeling and amyloid load. Hypertension was induced for 1 month by administration of l-NG-nitroarginine-methyl-ester in TgF344-AD rats at the prodromal stage. Following hypertension, nontransgenic rats showed transient cerebrovascular changes, whereas TgF344-AD animals exhibited sustained alterations in cerebrovascular function. Human umbilical cord perivascular cells in combination with scyllo-inositol, an inhibitor of Aβ oligomerization, resulted in normalization of hippocampal vascular function and remodeling, in contrast to either treatment alone. Prodromal stage hypertension exacerbates latter AD pathology, and the combination of human umbilical cord perivascular cells with amyloid clearance promotes cerebrovascular functional recovery.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Tina L Beckett
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Margaret M Koletar
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Mary E Hill
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Aaron Lai
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Arunachala Trivedi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Lynsie Thomason
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Adrienne Dorr
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | | | - Clifford L Librach
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Division of Obstetrics and Gynaecology, Laboratory Medicine and Pathobiology (C.L.), University of Toronto, Canada.,CReATe Research Program, Toronto, Canada (D.G., C.L.L.)
| | - Illsung L Joo
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - JoAnne McLaurin
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Bojana Stefanovic
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Department of Medical Biophysics (B.S.), University of Toronto, Canada
| |
Collapse
|
25
|
Collateral Vessels Have Unique Endothelial and Smooth Muscle Cell Phenotypes. Int J Mol Sci 2019; 20:ijms20153608. [PMID: 31344780 PMCID: PMC6695737 DOI: 10.3390/ijms20153608] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Collaterals are unique blood vessels present in the microcirculation of most tissues that, by cross-connecting a small fraction of the outer branches of adjacent arterial trees, provide alternate routes of perfusion. However, collaterals are especially susceptible to rarefaction caused by aging, other vascular risk factors, and mouse models of Alzheimer’s disease—a vulnerability attributed to the disturbed hemodynamic environment in the watershed regions where they reside. We examined the hypothesis that endothelial and smooth muscle cells (ECs and SMCs, respectively) of collaterals have specializations, distinct from those of similarly-sized nearby distal-most arterioles (DMAs) that maintain collateral integrity despite their continuous exposure to low and oscillatory/disturbed shear stress, high wall stress, and low blood oxygen. Examination of mouse brain revealed the following: Unlike the pro-inflammatory cobble-stoned morphology of ECs exposed to low/oscillatory shear stress elsewhere in the vasculature, collateral ECs are aligned with the vessel axis. Primary cilia, which sense shear stress, are present, unexpectedly, on ECs of collaterals and DMAs but are less abundant on collaterals. Unlike DMAs, collaterals are continuously invested with SMCs, have increased expression of Pycard, Ki67, Pdgfb, Angpt2, Dll4, Ephrinb2, and eNOS, and maintain expression of Klf2/4. Collaterals lack tortuosity when first formed during development, but tortuosity becomes evident within days after birth, progresses through middle age, and then declines—results consistent with the concept that collateral wall cells have a higher turnover rate than DMAs that favors proliferative senescence and collateral rarefaction. In conclusion, endothelial and SMCs of collaterals have morphologic and functional differences from those of nearby similarly sized arterioles. Future studies are required to determine if they represent specializations that counterbalance the disturbed hemodynamic, pro-inflammatory, and pro-proliferative environment in which collaterals reside and thus mitigate their risk factor-induced rarefaction.
Collapse
|
26
|
Mouse models of Alzheimer's disease cause rarefaction of pial collaterals and increased severity of ischemic stroke. Angiogenesis 2019; 22:263-279. [PMID: 30519973 PMCID: PMC6475514 DOI: 10.1007/s10456-018-9655-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/20/2018] [Indexed: 01/26/2023]
Abstract
Vascular dysfunction contributes to the progression and severity of Alzheimer's disease (AD). Patients with AD also sustain larger infarctions after ischemic stroke; however, the responsible mechanisms are unknown. Pial collaterals are the primary source of protection in stroke. Unfortunately, natural aging and other vascular risk factors cause a decline in collateral number and diameter (rarefaction) and an increase in stroke severity. Herein, we tested the hypothesis that AD accelerates age-induced collateral rarefaction and examined potential underlying mechanisms. Triple and double transgenic mouse models of AD both sustained collateral rarefaction by 8 months of age, well before the onset of rarefaction caused by aging alone (16 months of age). Rarefaction, which did not progress further at 18 months of age, was accompanied by a twofold increase in infarct volume after MCA occlusion. AD did not induce rarefaction of similarly sized pial arterioles or penetrating arterioles. Rarefaction was minimal and occurred only at 18 months of age in a parenchymal vascular amyloid-beta model of AD. Rarefaction was not associated with amyloid-beta deposition on collaterals or pial arteries, nor was plaque burden or CD11b+ cell density greater in brain underlying the collateral zones versus elsewhere. However, rarefaction was accompanied by increased markers of oxidative stress, inflammation, and aging of collateral endothelial and mural cells. Moreover, rarefaction was lessened by deletion of CX3CR1 and prevented by overexpression of eNOS. These findings demonstrate that mouse models of AD promote rarefaction of pial collaterals and implicate inflammation-induced accelerated aging of collateral wall cells. Strategies that reduce vascular inflammation and/or increase nitric oxide may preserve collateral function.
Collapse
|
27
|
Hanrahan JP, Wakefield JD, Wilson PJ, Mihova M, Chickering JG, Ruff D, Hall M, Milne GT, Currie MG, Profy AT. A Randomized, Placebo-Controlled, Multiple-Ascending-Dose Study to Assess the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of the Soluble Guanylate Cyclase Stimulator Praliciguat in Healthy Subjects. Clin Pharmacol Drug Dev 2018; 8:564-575. [PMID: 30422390 DOI: 10.1002/cpdd.627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/08/2018] [Indexed: 12/14/2022]
Abstract
Nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling is central to the regulation of several physiological processes, including blood flow and inflammation. Deficient NO signaling is implicated in multiple diseases. sGC stimulators are small molecules that enhance sGC activity, particularly in combination with NO. In a randomized, placebo-controlled phase 1 study, the safety, tolerability, pharmacokinetics, and pharmacodynamics of multiple ascending doses of the sGC stimulator praliciguat were assessed in 44 healthy adults. Four cohorts of 11 subjects (8 praliciguat, 3 placebo) received once-daily praliciguat for 14 days before up-titrating for 7 days (treatment sequences: 15/30 mg, 20/40 mg, 30/40 mg, and weight-based). All doses were tolerated. No serious or severe adverse events (AEs) were reported. The most common AEs in praliciguat recipients were headache and symptoms consistent with blood pressure (BP) lowering/vasodilation. There were no laboratory, vital sign, electrocardiographic, or platelet function findings indicative of a safety concern. Pharmacokinetics were dose proportional, with an effective half-life of 24-37 hours, supporting once-daily dosing. Praliciguat produced dose-related increases in plasma cGMP consistent with stimulation of sGC. Repeated once-daily dosing showed sustained decreases in BP. Results support evaluation of praliciguat for the treatment of conditions associated with deficient NO signaling.
Collapse
Affiliation(s)
| | | | | | | | | | - Dennis Ruff
- ICON Early Phase Services LLC, San Antonio, TX, USA
| | - Michael Hall
- Ironwood Pharmaceuticals Inc., Cambridge, MA, USA
| | - G Todd Milne
- Ironwood Pharmaceuticals Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
28
|
Wirth KJ. Role of Noradrenergic Brain Nuclei in the Regulation of Carotid Artery Blood Flow: Pharmacological Evidence from Anesthetized Pigs with Alpha-2 Adrenergic Receptor Modulator Drugs. J Alzheimers Dis 2018; 66:407-419. [DOI: 10.3233/jad-180340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Klaus J. Wirth
- Sanofi-Aventis Deutschland GmbH, R&D TA Immunology, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Thurgur H, Pinteaux E. Microglia in the Neurovascular Unit: Blood-Brain Barrier-microglia Interactions After Central Nervous System Disorders. Neuroscience 2018; 405:55-67. [PMID: 31007172 DOI: 10.1016/j.neuroscience.2018.06.046] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022]
Abstract
Over the past few decades, microglial cells have been regarded as the main executor of inflammation after acute and chronic central nervous system (CNS) disorders, responding rapidly to exogenous stimuli during acute trauma or infections, or signals released by cells undergoing cell death during conditions such as stroke, Alzheimer's disease (AD) and Parkinson's disease (PD). Barriers of the nervous system, and in particular the blood-brain barrier (BBB), play a key role in the normal physiological and cognitive functions of the brain. Being at the interface between the central and peripheral compartment, the BBB is regarded as a sensor of homeostasis, and any disruption within the brain or the systemic compartment triggers BBB dysfunction and neuroinflammation, both contributing to the pathogenesis of cerebrovascular disease. This involves a dynamic response mediated by all components of the neurovascular unit (NVU), and ongoing research suggests that BBB-microglia interaction is critical to dictate the microglial response to NVU injury. The present review aims to give an up-to-date account of the emerging critical role of BBB-microglia interactions during neuroinflammation, and how these could be targeted for the therapeutic treatment of major central inflammatory disease.
Collapse
Affiliation(s)
- Hannah Thurgur
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, United Kingdom.
| |
Collapse
|
30
|
Eguchi K, Shindo T, Ito K, Ogata T, Kurosawa R, Kagaya Y, Monma Y, Ichijo S, Kasukabe S, Miyata S, Yoshikawa T, Yanai K, Taki H, Kanai H, Osumi N, Shimokawa H. Whole-brain low-intensity pulsed ultrasound therapy markedly improves cognitive dysfunctions in mouse models of dementia - Crucial roles of endothelial nitric oxide synthase. Brain Stimul 2018; 11:959-973. [PMID: 29857968 DOI: 10.1016/j.brs.2018.05.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/01/2018] [Accepted: 05/20/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Therapeutic focused-ultrasound to the hippocampus has been reported to exert neuroprotective effects on dementia. In the present study, we examined whether the whole-brain LIPUS (low-intensity pulsed ultrasound) therapy is effective and safe in 2 mouse models of dementia (vascular dementia, VaD and Alzheimer's disease, AD), and if so, to elucidate the common underlying mechanism(s) involved. METHODS We used bilateral carotid artery stenosis (BCAS) model with micro-coils in male C57BL/6 mice as a VaD model and 5XFAD transgenic mice as an AD model. We applied the LIPUS therapy (1.875 MHz, 6.0 kHz, 32cycles) to the whole brain. RESULTS In both models, the LIPUS therapy markedly ameliorated cognitive impairments (Y-maze test and/or passive avoidance test) associated with improved cerebral blood flow (CBF). Mechanistically, the LIPUS therapy significantly increased CD31-positive endothelial cells and Olig2-positive oligodendrocyte precursor cells (OPCs) in the VaD model, while it reduced Iba-1-positive microglias and amyloid-β (Aβ) plaque in the AD model. In both models, endothelium-related genes were significantly upregulated in RNA-sequencing, and expressions of endothelial nitric oxide synthase (eNOS) and neurotrophins were upregulated in Western blotting. Interestingly, the increases in glia cells and neurotrophin expressions showed significant correlations with eNOS expression. Importantly, these beneficial effects of LIPUS were absent in eNOS-knockout mice. CONCLUSIONS These results indicate that the whole-brain LIPUS is an effective and non-invasive therapy for dementia by activating specific cells corresponding to each pathology, for which eNOS activation plays an important role as a common mechanism.
Collapse
Affiliation(s)
- Kumiko Eguchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiko Shindo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenta Ito
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsuyoshi Ogata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Kurosawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuta Kagaya
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuto Monma
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadamitsu Ichijo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sachie Kasukabe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University School of Medicine Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine Sendai, Japan
| | - Hirofumi Taki
- Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Hiroshi Kanai
- Department of Electronic Engineering, Tohoku University Graduate School of Engineering, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
31
|
Burrage E, Marshall KL, Santanam N, Chantler PD. Cerebrovascular dysfunction with stress and depression. Brain Circ 2018; 4:43-53. [PMID: 30276336 PMCID: PMC6126243 DOI: 10.4103/bc.bc_6_18] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Maintenance of adequate tissue perfusion through a dense network of cerebral microvessels is critical for the perseveration of normal brain function. Regulation of the cerebral blood flow has to ensure adequate delivery of nutrients and oxygen with moment-to-moment adjustments to avoid both hypo- and hyper-perfusion of the brain tissue. Even mild impairments of cerebral blood flow regulation can have significant implications on brain function. Evidence suggests that chronic stress and depression elicits multifaceted functional impairments to the cerebral microcirculation, which plays a critical role in brain health and the pathogenesis of stress-related cognitive impairment and cerebrovascular events. Identifying the functional and structural changes to the brain that are induced by stress is crucial for achieving a realistic understanding of how related illnesses, which are highly disabling and with a large economic cost, can be managed or reversed. This overview discusses the stress-induced alterations in neurovascular coupling with specific attention to cerebrovascular regulation (endothelial dependent and independent vasomotor function, microvessel density). The pathophysiological consequences of cerebral microvascular dysfunction with stress and depression are explored.
Collapse
Affiliation(s)
- Emily Burrage
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV, USA
| | - Kent L. Marshall
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Paul D. Chantler
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
32
|
D'Arrigo JS. Targeting Early Dementia: Using Lipid Cubic Phase Nanocarriers to Cross the Blood⁻Brain Barrier. Biomimetics (Basel) 2018; 3:E4. [PMID: 31105226 PMCID: PMC6352688 DOI: 10.3390/biomimetics3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, a frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease has been observed. Numerous published studies indicate that the preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic, which targets certain cell surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood⁻brain barrier. This targeting allows for various cell types related to Alzheimer's to be simultaneously searched out for localized drug treatment in vivo.
Collapse
|
33
|
Maternal high-salt diet alters redox state and mitochondrial function in newborn rat offspring's brain. Br J Nutr 2018; 119:1003-1011. [PMID: 29502538 DOI: 10.1017/s0007114518000235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Excessive salt intake is a common feature of Western dietary patterns, and has been associated with important metabolic changes including cerebral redox state imbalance. Considering that little is known about the effect on progeny of excessive salt intake during pregnancy, the present study investigated the effect of a high-salt diet during pregnancy and lactation on mitochondrial parameters and the redox state of the brains of resulting offspring. Adult female Wistar rats were divided into two dietary groups (n 20 rats/group): control standard chow (0·675 % NaCl) or high-salt chow (7·2 % NaCl), received throughout pregnancy and for 7 d after delivery. On postnatal day 7, the pups were euthanised and their cerebellum, hypothalamus, hippocampus, prefrontal and parietal cortices were dissected. Maternal high-salt diet reduced cerebellar mitochondrial mass and membrane potential, promoted an increase in reactive oxygen species allied to superoxide dismutase activation and decreased offspring cerebellar nitric oxide levels. A significant increase in hypothalamic nitric oxide levels and mitochondrial superoxide in the hippocampus and prefrontal cortex was observed in the maternal high-salt group. Antioxidant enzymes were differentially modulated by oxidant increases in each brain area studied. Taken together, our results suggest that a maternal high-salt diet during pregnancy and lactation programmes the brain metabolism of offspring, favouring impaired mitochondrial function and promoting an oxidative environment; this highlights the adverse effect of high-salt intake in the health state of the offspring.
Collapse
|
34
|
Zhang X, He X, Chen Q, Lu J, Rapposelli S, Pi R. A review on the hybrids of hydroxycinnamic acid as multi-target-directed ligands against Alzheimer’s disease. Bioorg Med Chem 2018; 26:543-550. [DOI: 10.1016/j.bmc.2017.12.042] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/14/2017] [Accepted: 12/24/2017] [Indexed: 01/31/2023]
|
35
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|
36
|
Alzheimer’s Disease, Brain Injury, and C.N.S. Nanotherapy in Humans: Sonoporation Augmenting Drug Targeting. Med Sci (Basel) 2017. [PMCID: PMC5753658 DOI: 10.3390/medsci5040029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Owing to the complexity of neurodegenerative diseases, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted nanoemulsion) are available. Versatile small-molecule drug(s) targeting multiple pathways of Alzheimer’s disease pathogenesis are known. By incorporating such drug(s) into the targeted lipid-coated microbubble/nanoparticle-derived (LCM/ND) lipid nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly scavenger receptor class B type I (SR-BI)), making it possible for various Alzheimer’s-related cell types to be simultaneously sought for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble (LCM) subpopulation (i.e., a stable LCM suspension); such LCM substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer’s patient.
Collapse
|
37
|
Cifuentes D, Poittevin M, Bonnin P, Ngkelo A, Kubis N, Merkulova-Rainon T, Lévy BI. Inactivation of Nitric Oxide Synthesis Exacerbates the Development of Alzheimer Disease Pathology in APPPS1 Mice (Amyloid Precursor Protein/Presenilin-1). Hypertension 2017; 70:613-623. [DOI: 10.1161/hypertensionaha.117.09742] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Diana Cifuentes
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Marine Poittevin
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Philippe Bonnin
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Anta Ngkelo
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Nathalie Kubis
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Tatyana Merkulova-Rainon
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| | - Bernard I. Lévy
- From the Institut des Vaisseaux et du Sang, Paris, France (M.P., A.N., T.M.-R., B.I.L.); INSERM U965, Paris, France (D.C., P.B., N.K., T.M.-R.); Université Paris Diderot, Sorbonne Paris Cité, France (P.B., N.K., B.I.L.); AP-HP, Hôpital Lariboisière, Paris, France (P.B., N.K.); and INSERM, U970, Paris, France (B.I.L.)
| |
Collapse
|
38
|
Angiotensin IV Receptors Mediate the Cognitive and Cerebrovascular Benefits of Losartan in a Mouse Model of Alzheimer's Disease. J Neurosci 2017; 37:5562-5573. [PMID: 28476949 DOI: 10.1523/jneurosci.0329-17.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/30/2017] [Accepted: 04/22/2017] [Indexed: 12/21/2022] Open
Abstract
The use of angiotensin receptor blockers (ARBs) correlates with reduced onset and progression of Alzheimer's disease (AD). The mechanism depicting how ARBs such as losartan restore cerebrovascular and cognitive deficits in AD is unknown. Here, we propose a mechanism underlying losartan's benefits by selectively blocking the effects of angiotensin IV (AngIV) at its receptor (AT4R) with divalinal in mice overexpressing the AD-related Swedish and Indiana mutations of the human amyloid precursor protein (APP mice) and WT mice. Young (3-month-old) mice were treated with losartan (∼10 mg/kg/d, 4 months), followed by intracerebroventricular administration of vehicle or divalinal in the final month of treatment. Spatial learning and memory were assessed using Morris water mazes at 3 and 4 months of losartan treatment. Cerebrovascular reactivity and whisker-evoked neurovascular coupling responses were measured at end point (∼7 months of age), together with biomarkers related to neuronal and vascular oxidative stress (superoxide dismutase-2), neuroinflammation (astroglial and microglial activation), neurogenesis (BrdU-labeled newborn cells), and amyloidosis [soluble amyloid-β (Aβ) species and Aβ plaque load]. Divalinal countered losartan's capacity to rescue spatial learning and memory and blocked losartan's benefits on dilatory function and baseline nitric oxide bioavailability. Divalinal reverted losartan's anti-inflammatory effects, but failed to modify losartan-mediated reductions in oxidative stress. Neither losartan nor divalinal affected arterial blood pressure or significantly altered the amyloid pathology in APP mice. Our findings identify activation of the AngIV/AT4R cascade as the underlying mechanism in losartan's benefits and a target that could restore Aβ-related cognitive and cerebrovascular deficits in AD.SIGNIFICANCE STATEMENT Antihypertensive medications that target the renin angiotensin system, such as angiotensin receptor blockers (ARBs), have been associated with lower incidence and progression of Alzheimer's disease (AD) in cohort studies. However, the manner by which ARBs mediate their beneficial effects is unknown. Here, the angiotensin IV receptor (AT4R) was identified as mediating the cognitive and cerebrovascular rescue of losartan, a commonly prescribed ARB, in a mouse model of AD. The AT4R was further implicated in mediating anti-inflammatory benefits. AT4R-mediated effects were independent from changes in blood pressure, amyloidosis, and oxidative stress. Overall, our results implicate the angiotensin IV/AT4R cascade as a promising candidate for AD intervention.
Collapse
|
39
|
Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol 2017; 312:H1-H20. [PMID: 27793855 PMCID: PMC5283909 DOI: 10.1152/ajpheart.00581.2016] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/10/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022]
Abstract
Increasing evidence from epidemiological, clinical and experimental studies indicate that age-related cerebromicrovascular dysfunction and microcirculatory damage play critical roles in the pathogenesis of many types of dementia in the elderly, including Alzheimer's disease. Understanding and targeting the age-related pathophysiological mechanisms that underlie vascular contributions to cognitive impairment and dementia (VCID) are expected to have a major role in preserving brain health in older individuals. Maintenance of cerebral perfusion, protecting the microcirculation from high pressure-induced damage and moment-to-moment adjustment of regional oxygen and nutrient supply to changes in demand are prerequisites for the prevention of cerebral ischemia and neuronal dysfunction. This overview discusses age-related alterations in three main regulatory paradigms involved in the regulation of cerebral blood flow (CBF): cerebral autoregulation/myogenic constriction, endothelium-dependent vasomotor function, and neurovascular coupling responses responsible for functional hyperemia. The pathophysiological consequences of cerebral microvascular dysregulation in aging are explored, including blood-brain barrier disruption, neuroinflammation, exacerbation of neurodegeneration, development of cerebral microhemorrhages, microvascular rarefaction, and ischemic neuronal dysfunction and damage. Due to the widespread attention that VCID has captured in recent years, the evidence for the causal role of cerebral microvascular dysregulation in cognitive decline is critically examined.
Collapse
Affiliation(s)
- Peter Toth
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Pecs, Hungary; and
| | - Stefano Tarantini
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anna Csiszar
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma;
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
40
|
Affiliation(s)
- Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine
| |
Collapse
|