1
|
Chen G, Xi E, Gu X, Wang H, Tang Q. The study on cuproptosis in Alzheimer's disease based on the cuproptosis key gene FDX1. Front Aging Neurosci 2024; 16:1480332. [PMID: 39759399 PMCID: PMC11696982 DOI: 10.3389/fnagi.2024.1480332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory and cognitive impairments. Previous studies have shown neuronal death in the brains of AD patients, but the role of cuproptosis and its associated genes in AD neurons remains unclear. Methods Intersection analysis was conducted using the AD transcriptome dataset GSE63060, neuron dataset GSE147528, and reported cuproptosis-related genes to identify the cuproptosis key gene FDX1 highly expressed in AD. Subsequently, cell experiments were performed by treating SH-SY5Y cells with Aβ25-35 to establish AD cell model. The real-time reverse transcriptase-polymerase chain reaction (RT-qPCR) and western blotting (WB) assays were employed to detect the expression levels of FDX1, DLAT, and DLST. Cell proliferation was analyzed by counting Kit-8 (CCK8), mitochondrial ROS levels were analyzed using flow cytometry. shRNA was used to downregulate FDX1 expression, followed by repetition of the aforementioned experiments. Clinical experiments utilized qPCR to detect FDX1 mRNA levels in peripheral venous blood of patients, and analyzed FDX1 expression differences in different APOE genotypes of AD patients. Finally, a protein-protein interaction (PPI) network of FDX1 was constructed based on the GeneMANIA database, immune infiltration analysis was conducted using R language, and transcription factors prediction for FDX1 was performed based on the ENCODE database. Results The cuproptosis key gene FDX1 showed significantly higher expression in peripheral blood and neuron models of AD compared to non-AD individuals, with significantly higher expression in APOE ε4/ε4 genotype than other APOE genotype of AD patients. Knockdown of FDX1 expression reduced the lipidation levels of DLAT and DLST in neurons, alleviated ROS accumulation in mitochondria, improved cell viability, and mitigated cuproptosis. Immune infiltration analysis results indicated a high enrichment of peripheral blood γδ-T lymphocytes in AD, and FDX1 was significantly associated with the infiltration of four immune cells and may be regulated by three transcription factors. Conclusion The cuproptosis key gene FDX1 is highly expressed in AD and may promote cuproptosis in AD neurons by regulating the lipidation levels of DLAT and DLST, thereby participating in the onset and development of AD. This provides a potential target for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Guilin Chen
- Department of Neurology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Erwei Xi
- Department of Neurology, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiaozhen Gu
- Institute of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Huili Wang
- Institute of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
2
|
Juul Rasmussen I, Luo J, Frikke-Schmidt R. Lipids, lipoproteins, and apolipoproteins: Associations with cognition and dementia. Atherosclerosis 2024; 398:118614. [PMID: 39340935 DOI: 10.1016/j.atherosclerosis.2024.118614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Due to increasing lifespan and aging populations globally there has been a steep rise in late-life dementia, which is now the second most common cause of death in high-income countries. In general, dementia can be divided into two major groups: Alzheimer's disease (AD) and vascular-related dementia (VD). AD is pathologically characterised by senile plaques containing amyloid-β and neurofibrillary tangles composed of hyperphosphorylated tau, whereas VD is dominated by vascular pathology such as cerebral small vessel disease, major strokes, and white matter lesions. Recently, the importance of vascular components in AD is increasingly recognized and it is estimated that up to 45 % of all dementia cases can be prevented by preventing or treating midlife cardiovascular risk factors such as physical inactivity, diabetes, and hypertension. Even though the brain contains approximately 25 % of the total body cholesterol pool, and several genetic variants related to the lipid metabolism have been identified in genome-wide associations studies of AD, the role of lipids, lipoproteins, and apolipoproteins in dementia risk is less well-known. In this review, we go through the current literature on lipids, lipoproteins, and apolipoproteins and risk of dementia. We conclude that the evidence is primarily insufficient or conflicting, possibly due to nonoptimal study designs. The future calls for large, prospective studies of midlife measurements of lipids, lipoproteins, and apolipoproteins and one-sample, individual level data Mendelian randomization studies to overcome survival bias. However, the current literature suggests that it is safe to say that what is good for the heart is good for the brain.
Collapse
Affiliation(s)
- Ida Juul Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 1, DK-2730, Herlev, Denmark.
| | - Jiao Luo
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 1, DK-2730, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| |
Collapse
|
3
|
Yan X, Ma Y, Yan L, Li S, Xu Y. Neuropeptides as Potential Biomarkers in Vascular Dementia. J Integr Neurosci 2024; 23:66. [PMID: 38538218 DOI: 10.31083/j.jin2303066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 07/15/2024] Open
Abstract
Neuropeptides are endogenous active substances within the central and peripheral nervous systems that play important roles in a wide range of brain functions, including metabolism, food intake, social behavior, reproduction, learning, sleep, and wakefulness. This article reviews recent advances in the involvement of neuropeptides in vascular dementia. Neuropeptides are present in the brain as chemical signals and last for nearly 50 years. Peptide hormones are chemical signals of the endocrine system. Thus, neuropeptides are the most diverse class of signaling molecules in the brain, involving the genomes of many mammals, encoding neuropeptide precursors and many bioactive neuropeptides. Here the aim is to describe the recent advances in classical neuropeptides, as well as putative neuropeptides from other families, in the control of or as diagnostic tools for vascular dementia. Additionally, its molecular mechanisms are described to explore new avenues of treatment and early diagnosis, as there is increasing evidence that dysregulation of vascular processes is associated with different pathological conditions.
Collapse
Affiliation(s)
- Xue Yan
- Department of Traditional Chinese Medicine, Haikou Maternal and Child Health Hospital, 570102 Haikou, Hainan, China
| | - Yihong Ma
- Department of Rehabilitation, The Second Affiliated Hospital of Hainan Medical University, 570311 Haikou, Hainan, China
| | - Limin Yan
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, 570311 Haikou, Hainan, China
| | - Simin Li
- Stomatological Hospital, Southern Medical University, 510280 Guangzhou, Guangdong, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, 271000 Taian, Shandong, China
| |
Collapse
|
4
|
Hosoki S, Hansra GK, Jayasena T, Poljak A, Mather KA, Catts VS, Rust R, Sagare A, Kovacic JC, Brodtmann A, Wallin A, Zlokovic BV, Ihara M, Sachdev PS. Molecular biomarkers for vascular cognitive impairment and dementia. Nat Rev Neurol 2023; 19:737-753. [PMID: 37957261 DOI: 10.1038/s41582-023-00884-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/15/2023]
Abstract
As disease-specific interventions for dementia are being developed, the ability to identify the underlying pathology and dementia subtypes is increasingly important. Vascular cognitive impairment and dementia (VCID) is the second most common cause of dementia after Alzheimer disease, but progress in identifying molecular biomarkers for accurate diagnosis of VCID has been relatively limited. In this Review, we examine the roles of large and small vessel disease in VCID, considering the underlying pathophysiological processes that lead to vascular brain injury, including atherosclerosis, arteriolosclerosis, ischaemic injury, haemorrhage, hypoperfusion, endothelial dysfunction, blood-brain barrier breakdown, inflammation, oxidative stress, hypoxia, and neuronal and glial degeneration. We consider the key molecules in these processes, including proteins and peptides, metabolites, lipids and circulating RNA, and consider their potential as molecular biomarkers alone and in combination. We also discuss the challenges in translating the promise of these biomarkers into clinical application.
Collapse
Affiliation(s)
- Satoshi Hosoki
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Gurpreet K Hansra
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Vibeke S Catts
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Ruslan Rust
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Amy Brodtmann
- Department of Neurology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Tzekaki EE, Tsolaki M, Pantazaki ΑA, Geromichalos G, Lazarou E, Kozori M, Sinakos Z. The pleiotropic beneficial intervention of olive oil intake on the Alzheimer's disease onset via fibrinolytic system. Exp Gerontol 2021; 150:111344. [PMID: 33836262 DOI: 10.1016/j.exger.2021.111344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022]
Abstract
The daily consumption of Extra Virgin Olive Oil (EVOO) in Mediterranean nutrition is tightly associated with lower frequency of many diseases' appearance, including Alzheimer's disease (AD). Fibrinolytic system is already assumed to be involved in AD pathophysiology through various factors, especially plasminogen activator inhibitor-1 (PAI-1), a2-antiplasmin (α2ΑP) and tissue plasminogen activator (tPA). We, here, present a biochemical study, as a continuation of a clinical trial of a cohort of 84 participants, focusing on the pleiotropic effect of the annual EVOO consumption on the fibrinolytic factors of Mild Cognitive Impairment (MCI) patients. The levels of all these fibrinolytic factors, measured by Enzyme-Linked Immunosorbent Assay (ELISA) method, were reduced in the serum of MCI patients annually administered with EVOO, versus not treated MCI patients, as well as AD patients. The well-established AD hallmarks (Aβ1-40 and Aβ1-42 species, tau, and p-tau) of MCI patients' group, annually administered with EVOO, were restored to levels equal to those of the cognitively-healthy group; in contrast to those patients not being administered, and their AD hallmarks levels increased at the end of the year. Moreover, one of the EVOO annual consumption multimodal effects on the MCI patients focused on the levels of an oxidative stress trademark, malondialdehyde (MDA), which displayed also a visible quenching; On the other hand, an increase exhibited in the MCI patients not consuming EVOO one year after, was attributed to the lack of the EVOO anti-oxidative properties. These outcomes are exploitable towards the establishment of natural products like EVOO, as a preventive remedy fighting this neurodegenerative disorder, AD. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT03362996 MICOIL gov Identifier: NCT03362996.
Collapse
Affiliation(s)
- Elena E Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Magda Tsolaki
- 1(st) Department of Neurology, Medical School, "AHEPA" General Hospital Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, 54124 Thessaloniki, Makedonia, Greece; Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece.
| | - Αnastasia A Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece.
| | - George Geromichalos
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Eftychia Lazarou
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Mahi Kozori
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Zacharias Sinakos
- Emeritus Professor of Hematology, Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, Greece
| |
Collapse
|
6
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
7
|
Yu QQ, Cheng DX, Xu LR, Li YK, Zheng XY, Liu Y, Li YF, Liu HL, Bai L, Wang R, Fan JL, Liu EQ, Zhao SH. Urotensin II and urantide exert opposite effects on the cellular components of atherosclerotic plaque in hypercholesterolemic rabbits. Acta Pharmacol Sin 2020; 41:546-553. [PMID: 31685976 PMCID: PMC7468446 DOI: 10.1038/s41401-019-0315-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022]
Abstract
Increasing levels of plasma urotensin II (UII) are positively associated with atherosclerosis. In this study we investigated the role of macrophage-secreted UII in atherosclerosis progression, and evaluated the therapeutic value of urantide, a potent competitive UII receptor antagonist, in atherosclerosis treatment. Macrophage-specific human UII-transgenic rabbits and their nontransgenic littermates were fed a high cholesterol diet for 16 weeks to induce atherosclerosis. Immunohistochemical staining of the cellular components (macrophages and smooth muscle cells) of aortic atherosclerotic lesions revealed a significant increase (52%) in the macrophage-positive area in only male transgenic rabbits compared with that in the nontransgenic littermates. However, both male and female transgenic rabbits showed a significant decrease (45% in males and 31% in females) in the smooth muscle cell-positive area compared with that of their control littermates. The effects of macrophage-secreted UII on the plaque cellular components were independent of plasma lipid level. Meanwhile the wild-type rabbits were continuously subcutaneously infused with urantide (5.4 µg· kg-1· h-1) using osmotic mini-pumps. Infusion of urantide exerted effects opposite to those caused by UII, as it significantly decreased the macrophage-positive area in male wild-type rabbits compared with that of control rabbits. In cultured human umbilical vein endothelial cells, treatment with UII dose-dependently increased the expression of the adhesion molecules VCAM-1 and ICAM-1, and this effect was partially reversed by urantide. The current study provides direct evidence that macrophage-secreted UII plays a key role in atherogenesis. Targeting UII with urantide may promote plaque stability by decreasing macrophage-derived foam cell formation, which is an indicator of unstable plaque.
Collapse
|
8
|
Tang Q, Wang F, Yang J, Peng H, Li Y, Li B, Wang S. Revealing a Novel Landscape of the Association Between Blood Lipid Levels and Alzheimer's Disease: A Meta-Analysis of a Case-Control Study. Front Aging Neurosci 2020; 11:370. [PMID: 32116643 PMCID: PMC7025526 DOI: 10.3389/fnagi.2019.00370] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
Objectives: Blood lipid profiles have been ambiguously reported as biomarkers of AD in recent years. This study was conducted to evaluate the correlation between blood lipid levels and AD in later-life and to explore the effectiveness and reliability of blood lipid profiles as biomarkers of AD. Methods: Database searching was conducted using PubMed, the Cochrane Library, EMBASE, and Medline. This study was designed following the Meta-analysis of Observational Studies in Epidemiology (MOOSE) criteria. Review Manager 5.3 (RevMan 5.3) software was adopted to perform meta-analysis evaluating the standard mean difference (SMD) with its 95% confidence intervals (CI). Results: A total of 5,286 participants were enrolled from 27 case-control studies in this meta-analysis. The pooled results demonstrated that total cholesterol (TC) level was significantly associated with AD in late-life (SMD = 0.17, 95% CI: [0.01, 0.32], P = 0.03), especially in the subgroup under 70 years old (SMD: 0.45, 95% CI: [0.11, 0.79], P = 0.01) and the subgroup of Western population (SMD: 0.29, 95% CI: [0.04, 0.53], P = 0.02). In the subgroup under 70 years old, the high-density lipoprotein cholesterol (HDL-C) level (SMD = -0.50, 95% CI: [-0.76, -0.25], P = 0.0001) and the low-density lipoprotein cholesterol (LDL-C) level (SMD = 0.59, 95% CI: [0.02, 1.16], P = 0.04) in the AD group were significantly lower and higher than in the control group, respectively. In the subgroup with a sample size larger than 100 subjects, the LDL-C level was significantly higher in AD patients than in the control elderly group (SMD = 0.31, 95% CI: [0.05, 0.56], P = 0.02). There was no significant association between triglyceride (TG) levels and AD in later-life (SMD = -0.00, 95% CI: [-0.12, 0.12], P = 1.00). Conclusion: TC can be a new predictive biomarker of AD or cognitive decline in later-life. Increased TC levels are found to be associated with an elevated risk of AD. Decreased HDL-C levels and increased LDL-C levels may relate to an elevated risk of AD in subjects aged 60-70. Further comprehensive researches will be necessary in the future.
Collapse
Affiliation(s)
- Qianyun Tang
- Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Nursing, Central South University, Changsha, China
| | - Fengling Wang
- Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Nursing, Central South University, Changsha, China
| | - Jingjing Yang
- Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Nursing, Central South University, Changsha, China
| | - Hua Peng
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Geriatric Department, Xiangya Hospital, Central South University, Changsha, China
| | - Shuhong Wang
- Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Nursing, Central South University, Changsha, China
| |
Collapse
|
9
|
Zhou Z, Liang Y, Zhang X, Xu J, Lin J, Zhang R, Kang K, Liu C, Zhao C, Zhao M. Low-Density Lipoprotein Cholesterol and Alzheimer's Disease: A Systematic Review and Meta-Analysis. Front Aging Neurosci 2020; 12:5. [PMID: 32082137 PMCID: PMC7002548 DOI: 10.3389/fnagi.2020.00005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: To assess the association between low-density lipoprotein cholesterol (LDL-c) and risk of Alzheimer's disease (AD). Methods: Embase, Pubmed, and Web of Science were searched until June 2019. Standard mean difference (SMD) with 95% confidence intervals (CI) was estimated using random-effects models. Results: Our meta-analysis of 26 studies revealed higher levels of LDL-c in AD than that of non-dementia controls (SMD = 0.35, 95% CI 0.12-0.58, p < 0.01). The meta-regression analysis on confounders showed that age (p < 0.01, Adj R-squared = 92.41%) and cardiovascular disease (p = 0.01, Adj R-squared = 85.21%), but not the body mass index, education, smoking, hypertension and diabetes mellitus, exerted an impact on the relationship between LDL-c and risk of ICH. Further subgroup analysis of age showed LDL-c levels in AD patients aged 60-70 were higher than that of non-dementia (60 ≤ age < 70: SMD = 0.80, 95% CI 0.23-1.37, p < 0.01); but no association between the SMD of AD in LDL-c and age over 70 was noted across the studies (70 ≤ age < 77: SMD = -0.02, 95% CI -0.39~0.34, p = 9.0; 77 ≤ age < 80: SMD = 0.15, 95% CI -0.17~0.47, p = 0.35; ≥80: SMD = 0.53, 95% CI -0.04~1.11, p = 0.07). The concentrations of LDL-c during the quintile interval of 3~4 were positively associated with AD (121 ≤ concentration < 137: SMD = 0.98, 95% CI 0.13~1.82, p = 0.02; ≥137: SMD = 0.62, 95% CI 0.18~1.06, p < 0.01); whereas there was no correlation between AD and LDL-c within the quintile interval of 1~2 (103.9 ≤ concentration < 112: SMD = 0.08, 95% CI -0.20~0.35, p = 0.59; 112 ≤ concentration < 121: SMD = -0.26, 95% CI -0.58~0.06, p = 0.11). Conclusions: Elevated concentration of LDL-c (>121 mg/dl) may be a potential risk factor for AD. This association is strong in patients aged 60-70 years, but vanishes with advancing age.
Collapse
Affiliation(s)
- Zhike Zhou
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yifan Liang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoqian Zhang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Junjie Xu
- Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jueying Lin
- Department of Emergency, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Rongwei Zhang
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Kexin Kang
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chang Liu
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chuansheng Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Mei Zhao
- Department of Cardiology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
10
|
Zhou F, Chen S. Effects of Gender and Other Confounding Factors on Leptin Concentrations in Alzheimer's Disease: Evidence from the Combined Analysis of 27 Case-Control Studies. J Alzheimers Dis 2019; 62:477-486. [PMID: 29439354 DOI: 10.3233/jad-170983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Leptin, as a link between fat mass and the brain, has been reported to be associated with gender. The gender differences in leptin levels between Alzheimer's disease (AD) and healthy elderly controls are inconclusive so far. To quantitatively summarize the leptin data available from female and male patients with AD, we searched PubMed and EMBASE for articles published from inception to July 20, 2017. Data were extracted from 27 studies, consisting of 3,014 participants. The pooled results showed that the overall leptin levels were lower in AD (Hedges' g = -0.481; p = 0.002) than in controls, and the leptin levels in whole blood and serum were decreased with moderate and large effect sizes (g = -0.677, -0.839; respectively; both of p-values <0.001) in AD compared with controls. In blood, there were significantly lower concentrations of leptin in female AD than in female controls (g = -0.590; p = 0.014), but not in male case-control group (g = -0.666; p = 0.067). Meta-regression analysis demonstrated that the decreased extent of leptin levels in AD paralleled the degree of the severity of dementia symptoms, as well as the alterations of body mass index (p-values ≤0.002). The findings provide strong evidence that 1) the blood concentrations of leptin are lower in female AD patients than in female controls; and 2) the greater the severity of dementia symptoms, the greater the decreases in the blood leptin levels. But more future investigations on the blood leptin levels in male AD patients is warranted.
Collapse
Affiliation(s)
- Futao Zhou
- College of Medicine and Health, Lishui University, Lishui Zhejiang, China
| | - Shuangrong Chen
- College of Engineering, Lishui University, Lishui Zhejiang, China
| |
Collapse
|
11
|
Wu Y, Wang Z, Jia X, Zhang H, Zhang H, Li J, Zhang K. Prediction of Alzheimer's disease with serum lipid levels in Asian individuals: a meta-analysis. Biomarkers 2019; 24:341-351. [PMID: 30663433 DOI: 10.1080/1354750x.2019.1571633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background: The serum lipid profile has become a routine clinical test and used as an important predictor for Alzheimer's disease (AD), although its predictive value remains undetermined. Objective: To evaluate the role of serum lipid levels in predicting the risk of AD. Methods: Meta-analyses were conducted using Comprehensive Meta-analyses (CMA) software to investigate the association between four conventional serum lipid profile parameters and the risk of AD, focused on samples from Asian. Results: In total, 3423 AD patients and 6127 healthy participants were involved. The results demonstrated that AD patients showed higher LDL-C and TC levels (SMD = 0.27, 95% CI: 0.04-0.51, p = 0.02 for LDL-C; SMD = 0.25, 95% CI: 0.05-0.46, p = 0.02 for TC) compared with those of healthy controls. People with higher LDL-C and/or TC levels had an increased risk of AD (OR = 1.64, 95% CI: 1.07-2.51 for LDL-C and OR = 1.58, 95% CI: 1.10-2.92 for TC). Conclusions: This study provided evidence that serum LDL-C and TC levels were associated with the risk of AD in Asian individuals. The routine lipid profile may be useful for AD diagnosis, monitoring and treatment.
Collapse
Affiliation(s)
- Yufei Wu
- a College of Life Science, Institute of Health & Population Northwest University , Xi'an , China
| | - Zhibin Wang
- a College of Life Science, Institute of Health & Population Northwest University , Xi'an , China
| | - Xueping Jia
- a College of Life Science, Institute of Health & Population Northwest University , Xi'an , China
| | - Huan Zhang
- b The 2nd Affiliated Hospital , Xi'an Jiaotong University , Xi'an , China
| | - Hong Zhang
- c Neurology Department , Shaanxi Provincial People's Hospital , Xi'an , China
| | - Junlin Li
- a College of Life Science, Institute of Health & Population Northwest University , Xi'an , China.,d Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education) , Northwest University , Xi'an , China
| | - Kejin Zhang
- a College of Life Science, Institute of Health & Population Northwest University , Xi'an , China.,d Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education) , Northwest University , Xi'an , China
| |
Collapse
|
12
|
Sun SL, Liu LM. Urotensin II: an inflammatory cytokine. J Endocrinol 2019; 240:JOE-18-0505.R2. [PMID: 30601760 DOI: 10.1530/joe-18-0505] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022]
Abstract
Urotensin II (UII) is a polypeptide molecule with neurohormone-like activity. It has been confirmed that UII is widely distributed in numerous organs of different animal species from fish to mammals, including humans. The UII receptor is orphan G-protein coupled receptor 14, also known as UT. The tissue distribution of UII and UT is highly consistent, and their expression may be regulated by autocrine and paracrine mechanisms. In the body, UII has many physiological and pathophysiological activities, such as vasoconstrictor and vasodilatory actions, cell proliferation, pro-fibrosis, neuroendocrine activity, insulin resistance, and carcinogenic and inflammatory effects, which have been recognized only in recent years. In fact, UII is involved in the process of inflammatory injury and plays a key role in the onset and development of inflammatory diseases. In this paper, we will review the roles UII plays in inflammatory diseases.
Collapse
Affiliation(s)
- Sui-Lin Sun
- S Sun, Department of Infection, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China, Nanchang, China
| | - Liang-Ming Liu
- L Liu, Department of Infection, Songjiang Hospital Affiliated to First People's Hospital, Shanghai Jiaotong University, Shanghai, 201600, China
| |
Collapse
|
13
|
Liu W, Han Q, Liu Q, Liang G, Wang J, Liu C. An investigation into the expression and mechanism of action of urotensin II in chronic pressure-overloaded rat hearts. Mol Med Rep 2015; 12:6626-34. [PMID: 26323194 PMCID: PMC4626172 DOI: 10.3892/mmr.2015.4244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to investigate the role and mode of action of urotensin II (U II) in the occurrence and progression of cardiac fibrosis in a pressure-overload rat model. Coarctation of the abdominal aorta was used to establish an animal model, and postoperative echocardiography, hemodynamic detection, hematoxylin and eosin staining, Masson staining and immunohistochemistry were performed to assess the changes in cardiac function and pathology. The expression levels of U II, G-protein-coupled receptor 14 and collagen (Col) I and Col III in the myocardial tissues were also analyzed. Neonatal rat fibroblasts were isolated, cultured and subsequently, generations 3–5 were randomly divided into different groups for the detection of Col I and Col III levels by enzyme-linked immunosorbent assay and western blotting. The protein expression levels were markedly increased in the model group, and this increase correlated with an increase in myocardial fibrosis. In cultured neonatal rat fibroblast cells, 10−8 mol/l U II significantly stimulated the synthesis of Col I and Col III (P<0.01) compared with the control group. Compared with the U II group, the administration of KT5720 (1 mol/l) or SB-611812 (1 mol/l) significantly reduced the synthesis and expression levels of Col I and Col III (P<0.05). U II may exert an important role in the process of myocardial fibrosis in chronic pressure-overload rats, and the cyclic adenosine monophosphate-protein kinase A signaling pathway may be partly involved in this process.
Collapse
Affiliation(s)
- Wenyuan Liu
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Qinghua Han
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Qinghua Liu
- Teaching and Research Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Gang Liang
- Department of Pathology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jin Wang
- Teaching and Research Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Chengfang Liu
- Teaching and Research Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
14
|
Oh J, Lee HJ, Song JH, Park SI, Kim H. Plasminogen activator inhibitor-1 as an early potential diagnostic marker for Alzheimer's disease. Exp Gerontol 2014; 60:87-91. [PMID: 25304332 DOI: 10.1016/j.exger.2014.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/29/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in individuals over 65 years old. However, to date, no useful early diagnostic markers for AD have been discovered. We examined the utility of plasminogen activator inhibitor-1 (PAI-1) as a potential biomarker for AD in subjects with mild cognitive impairment (MCI) or AD, as well as in nondemented healthy controls. Plasma PAI-1 levels were measured by enzyme-linked immunosorbent assays (ELISAs) in samples collected from 76 patients with MCI, 74 patients with AD, and 76 healthy controls. Our results show that plasma PAI-1 levels gradually increased as dementia progressed. The mean levels of plasma PAI-1 in patients with MCI and AD patients were significantly higher than those of in healthy controls. Consistently, neuropsychological examination (e.g., MMSE, CDR) also demonstrated significant correlations between the plasma PAI-1 levels and cognitive function. In conclusion, the level of plasma PAI-1 is a potential biomarker for the early detection and diagnosis of AD.
Collapse
Affiliation(s)
- Jaeho Oh
- Division of Brain Diseases, Center for Biomedical Science, Korea National Institute of Health, Osong, Chungcheongbuk-do 361-951, South Korea
| | - Hye-Ja Lee
- Division of Metabolic Diseases, Center for Biomedical Science, Korea National Institute of Health, Osong, Chungcheongbuk-do 361-951, South Korea
| | - Ji-Hyun Song
- Division of Brain Diseases, Center for Biomedical Science, Korea National Institute of Health, Osong, Chungcheongbuk-do 361-951, South Korea
| | - Sang Ick Park
- Division of Brain Diseases, Center for Biomedical Science, Korea National Institute of Health, Osong, Chungcheongbuk-do 361-951, South Korea.
| | - Hyunyoung Kim
- Division of Brain Diseases, Center for Biomedical Science, Korea National Institute of Health, Osong, Chungcheongbuk-do 361-951, South Korea.
| |
Collapse
|
15
|
Zengin H, Erbay AR, Okuyucu A, Alaçam H, Yüksel S, Meriç M, Soylu K, Gedikli Ö, Murat N, Gülel O, Demircan S, Akın F, Yılmaz Ö, Şahin M. The relationship between coronary slow flow phenomenon and urotensin-II: A prospective and controlled study. Anatol J Cardiol 2014; 15:475-9. [PMID: 25430414 PMCID: PMC5779140 DOI: 10.5152/akd.2014.5481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The underlying mechanism of coronary slow flow (CSF) has not yet been clarified, although many studies have been conducted to understand its pathophysiology. In this study, we investigated the role of a very potent vasoconstrictor, urotensin-II (UII), in the pathophysiology of CSF. This prospective and controlled investigation aimed to evaluate the association between CSF and serum levels of UII. METHODS Our study included 32 patients with slow flow in any coronary artery and 32 patients with normal coronary arteries. Coronary flow was calculated using the Thrombolysis in Myocardial Infarction (TIMI) frame count (TFC) method, and CSF was defined as TFC ≥39 for the left anterior descending artery, TFC ≥27 for the circumflex coronary artery, and TFC ≥24 for the right coronary artery. UII levels in blood samples obtained from both groups were measured by enzyme-linked immunosorbent assay (ELISA) method. RESULTS UII levels were significantly higher in the CSF group than in the control group [122 pg/mL (71-831), 95 pg/mL (21-635), respectively; p<0.001]. High-density lipoprotein (HDL) levels were lower in the CSF group, and leukocyte counts were significantly higher. A positive correlation between UII and mean TFC (r=0.524, p=0.002) was found in the CSF group. The multivariate logistic regression analysis determined that UII, HDL, and cigarette smoking were independent indicators in predicting CSF (OR=1.010, 95% confidence interval 1.002-1014, p=0.019; OR=0.927, 95% confidence interval 0.869-0.988, p=0.019; OR=5.755, 95% confidence interval 1.272-26.041, p=0.021, respectively). CONCLUSION Serum UII levels were found to be significantly higher in the CSF group, suggesting that UII may be one of the underlying factors in the pathogenesis of CSF.
Collapse
Affiliation(s)
- Halit Zengin
- Department of Cardiology, Faculty of Medicine, Ondokuz Mayıs University; Samsun-Turkey.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Association between human urotensin II and essential hypertension--a 1:1 matched case-control study. PLoS One 2013; 8:e81764. [PMID: 24339964 PMCID: PMC3858253 DOI: 10.1371/journal.pone.0081764] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/16/2013] [Indexed: 12/18/2022] Open
Abstract
Objective We aimed to evaluate the controversial association between human urotensin II and essential hypertension in untreated hypertensive cases and normotensive controls. Methods 197 newly diagnosed hypertensive patients and 197 age- and sex-matched normotensive controls were studied. Plasma urotensin II, nitric oxide metabolites, and other traditional biomarkers were examined. Results Hypertensive patients had higher urotensin II [median (interquartile rang): 9.32 (7.86–11.52) ng/mL vs 8.52 (7.07–10.41) ng/mL] and lower nitric oxide metabolites [19.19 (2.55–38.48) µmol/L vs 23.83 (11.97–43.40) µmol/L] than normotensive controls. Urotensin II was positively correlated with systolic blood pressure (r = 0.169, P<0.001) and diastolic blood pressure (r = 0.113, P = 0.024) while negatively correlated with nitric oxide metabolites (r = −0.112, P = 0.027). In multivariate regression analysis, subjects in the highest quartile of urotensin II were more likely to have hypertension than those in the lowest quartile (OR, 2.58; 95% CI, 1.21–5.49). Sub-group analyses in 106 pairs of cases and controls with either both normal or both abnormal nitric oxide metabolites levels showed that the association between urotensin II levels and hypertension persisted (P value for trend = 0.039). Conclusion Human urotensin II is markedly associated with essential hypertension, and the association is independent of nitric oxide metabolites. Our results indicated that urotensin II might be an independent risk factor for essential hypertension.
Collapse
|
17
|
You Z, Al Kindi H, Abdul-Karim A, Barrette PO, Schwertani A. Blocking the urotensin II receptor pathway ameliorates the metabolic syndrome and improves cardiac function in obese mice. FASEB J 2013; 28:1210-20. [PMID: 24297699 DOI: 10.1096/fj.13-236471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The metabolic syndrome is defined by the presence of hyperlipidemia, obesity, hypertension, and diabetes. The syndrome is associated with significant cardiovascular morbidity and mortality. The aim of the present study was to determine the role of the vasoactive peptide urotensin II (UII) in the pathogenesis of the metabolic syndrome. We used obese mice (ob/ob) to determine the effect of UII receptor (UT) blockage on the different facets of the metabolic syndrome with special emphasis on cardiac function. Our data demonstrate a significant increase in UII and UT expression in the myocardium of obese mice accompanied by a significant decrease in sarco/endoplasmic reticulum Ca(2+)-ATPase 2a (SERCA2a) expression, as well as intracellular Na(+) and Ca(2+) compared with wild-type mice (P<0.05). Treatment of ob/ob mice with the UII receptor antagonist SB657510 significantly improved glucose levels, blood pressure, hyperlipidemia, expression of myocardial SERCA2a, intracellular Na(+) and Ca(2+) and cardiac function in association with a decrease in weight gain, and mammalian target of rapamycin (mTOR) and sodium/hydrogen exchanger 1 (NHE-1) protein expression compared with vehicle (P<0.05). These findings demonstrate an important role for UII in the pathogenesis of the metabolic syndrome and suggest that the use of UT receptor antagonists may provide a new therapeutic tool for the treatment of this syndrome.
Collapse
Affiliation(s)
- Zhipeng You
- 1McGill University Health Center, Ste. C9-166, Montreal General Hospital, 1650 Cedar Ave., Montreal, Quebec H3G 1A4, Canada.
| | | | | | | | | |
Collapse
|
18
|
Serum lipoprotein-A levels in healthy subjects indicate a lurking cerebro- and cardio-vascular risk in the younger population. Brain Res Bull 2013; 97:48-52. [DOI: 10.1016/j.brainresbull.2013.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/16/2013] [Accepted: 05/21/2013] [Indexed: 11/20/2022]
|
19
|
Zhao J, Yu QX, Kong W, Gao HC, Sun B, Xie YQ, Ren LQ. The urotensin II receptor antagonist, urantide, protects against atherosclerosis in rats. Exp Ther Med 2013; 5:1765-1769. [PMID: 23837070 PMCID: PMC3702698 DOI: 10.3892/etm.2013.1052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/05/2013] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to explore the use of urantide as an antagonist of the urotensin II (UII) receptor, G protein-coupled receptor 14 (GPR14), to protect against atherosclerosis (AS) in rats. The AS rat model was induced by an intraperitoneal injection of vitamin D3 (VD3) into rats fed with a high-fat diet for four weeks. Urantide was then injected into the rats. Immunohistochemical staining, serum biochemical assay, reverse transcription-polymerase chain reaction (RT-PCR) and western blotting were used to investigate the expression of UII and its receptor GPR14 in the AS rat model. Four weeks after induction, pathological changes typical of AS were observed in the AS rat model. In the plaques of the aortic tunica intima and tunica media, expression of UII and GPR14 was observed. The protein and gene expression levels of UII and GPR14 in the model group were significantly increased compared with those in the normal group (P<0.01). Urantide ameliorated the pathological changes of AS in the rat model and reduced the gene and protein expression levels of UII and GPR14 (P<0.05 or P<0.01). UII is associated with AS and the UII receptor GPR14-specific antagonist, urantide, demonstrates the ability to protect against AS. Thus, this study provides new insight and experimental theories for the clinical application of urantide to treat AS.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000
| | | | | | | | | | | | | |
Collapse
|
20
|
Zhao J, Ding W, Song N, Dong X, Di B, Peng F, Tang C. Urotensin II-induced collagen synthesis in cultured smooth muscle cells from rat aortic media and a possible involvement of transforming growth factor-β1/Smad2/3 signaling pathway. ACTA ACUST UNITED AC 2013; 182:53-8. [PMID: 23403244 DOI: 10.1016/j.regpep.2012.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 11/22/2012] [Accepted: 12/17/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies suggest that urotensin II (UII) and transforming growth factor-β1 (TGF-β1) both have critical roles in vascular remodeling. UII is a recently discovered vasoconstrictive peptide that is involved in the pathogenesis of atherosclerosis, restenosis and hypertension. TGF-β1 is an important factor that has a pivotal role in vascular fibrosis. This study aimed to explore whether TGF-β1 is involved in UII-induced collagen synthesis in rat aortic vascular smooth muscle cells (VSMCs) and examined the effects and mechanisms of UII on collagen synthesis and secretion in VSMCs. METHODS VSMCs were prepared by the explant culture method. TGF-β1 and collagen I secretions from the cells were determined by enzyme-linked immunosorbent assay (ELISA). The mRNA and protein expressions of TGF-β1, collagen I, Smad2 and Smad3 were determined using Real-time RT-PCR and Western blotting. RESULTS UII dose-dependently promoted TGF-β1 protein expression and secretion from VSMCs, with maximal effect at 10(-8) mol/l at 24 h for protein expression and 10(-7) mol/l at 24 h for protein secretion (both P<0.01). Moreover, UII dose-dependently promoted Smad2 and Smad3 mRNA expression in VSMCs, with maximal effect at 10(-8) mol/l for 12 h (both P<0.01). The effects of UII were significantly inhibited by its receptor antagonists urantide (10(-6) mol/l) or SB-710411 (10(-6) mol/l), and by the mitogen-activated protein kinase (MAPK/ERK) inhibitor PD98059 (10(-6) mol/l). UII dose-dependently promoted collagen I mRNA expression and protein secretion in VSMCs, with maximal effect at 10(-8) mol/l at 12h for mRNA expression and 10(-6) mol/l at 24 h for protein secretion (both P<0.01). Collagen synthesis and secretion from VSMCs induced by UII were inhibited significantly by a TGF-β1-specific neutralizing antibody, SB-431542 (an antagonist of the TGF-β1 type II receptor) and PD98059 (all P<0.01). CONCLUSIONS This study suggests that UII could induce collagen synthesis and secretion through upregulation of TGF-β1 expression and secretion in VSMCs, and that TGF-β1/Smad2/3 signaling might be one of the important pathways by which UII is involved in vascular fibrosis.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Cardiology, Peking University First Hospital, PR China
| | | | | | | | | | | | | |
Collapse
|
21
|
You Z, Genest J, Barrette PO, Hafiane A, Behm DJ, D'Orleans-Juste P, Schwertani AG. Genetic and pharmacological manipulation of urotensin II ameliorate the metabolic and atherosclerosis sequalae in mice. Arterioscler Thromb Vasc Biol 2012; 32:1809-16. [PMID: 22723440 DOI: 10.1161/atvbaha.112.252973] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Urotensin II (UII) is a potent vasoactive peptide that binds to the urotensin receptor-coupled receptor-14 (known as UT) and exerts a wide range of actions in humans and experimental animals. We tested the hypothesis that UII gene deletion or UT blockade ameliorate experimental atherosclerosis. METHODS AND RESULTS We observed a significant reduction in weight gain, visceral fat, blood pressure, circulating plasma lipids, and proatherogenic cytokines and improvement of glucose tolerance in UII knockout mice compared with wild type (P<0.05). Deletion of UII after an apolipoprotein E knockout resulted in a significant reduction in serum cytokines, adipokines, and aortic atherosclerosis compared with apolipoprotein E knockout mice. Similarly, treatment of apolipoprotein E knockout mice fed on high-fat diet with the UT antagonist SB657510A reduced weight gain, visceral fat, and hyperlipidemia and improved glucose tolerance (P<0.05) and attenuated the initiation and progression of atherosclerosis. The UT antagonist also decreased aortic extracellular signal-regulated kinase 1/2 phosphorylation and oxidant formation and serum level of cytokines (P<0.05). CONCLUSIONS These findings demonstrate for the first time the role of UII gene deletion in atherosclerosis and suggest that the use of pharmaceutical agents aimed at blocking the UII pathway may provide a novel approach in the treatment of atherosclerosis and its associated precursors such as obesity, hyperlipidemia, diabetes mellitus, and hypertension.
Collapse
Affiliation(s)
- Zhipeng You
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Obesity and the ageing brain: could leptin play a role in neurodegeneration? Curr Gerontol Geriatr Res 2011; 2011:708154. [PMID: 22013440 PMCID: PMC3195276 DOI: 10.1155/2011/708154] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 08/15/2011] [Indexed: 01/19/2023] Open
Abstract
Obesity and ageing are both characteristics of the human population that are on the increase across the globe. It has long been established that ageing is the major risk factor for neurodegenerative conditions such as Alzheimer's disease, and it is becoming increasingly evident that obesity is another such factor. Leptin resistance or insensitivity has been uncovered as a cause of obesity, and in addition the leptin signalling system is less potent in the elderly. Taken together, these findings reveal that this molecule may be a link between neurodegeneration and obesity or ageing. It is now known that leptin has beneficial effects on both the survival and neurophysiology of the neurons that are lost in Alzheimer's disease suggesting that it may be an important research target in the quest for strategies to prevent, halt, or cure this condition.
Collapse
|
23
|
Kiss RS, You Z, Genest J, Behm DJ, Giaid A. Urotensin II differentially regulates macrophage and hepatic cholesterol homeostasis. Peptides 2011; 32:956-63. [PMID: 21376094 DOI: 10.1016/j.peptides.2011.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/21/2011] [Accepted: 02/22/2011] [Indexed: 12/12/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide with pleiotropic activity. Interestingly, UII levels are elevated in hyperlipidemic patients, and UII induces lipase activity in some species. However, the exact role UII plays in cholesterol homeostasis remains to be elucidated. UII knockout (UII KO) mice were generated and a plasma lipoprotein profile, and hepatocytes and macrophages cholesterol uptake, storage and synthesis was determined. UII KO had a decreased LDL cholesterol profile and liver steatosis compared to wildtype mice (WT). UII KO macrophages demonstrated enhanced ACAT activity and LDL uptake in the short term (up to 4h), of which more LDL-delivered exogenously derived cholesterol was incorporated into cholesteryl ester (CE) than the WT macrophages. UII KO macrophages generated more than two times the amount of de novo endogenously synthesized cholesterol, and of this cholesterol more than two times the relative amount was esterified to CE. In comparison, results in hepatocytes demonstrated that far more exogenously derived cholesterol was incorporated into CE in the WT cells, generating almost ten times the amount of CE than UII KO. WT cells synthesize de novo almost ten times the amount of cholesterol than UIIKO, and of that cholesterol, almost two times the amount of CE in WT than UII KO hepatocytes. In addition, more ApoB lipoproteins were secreted from WT than UII KO hepatocytes. These results demonstrate a fundamental difference between macrophages and hepatocytes in terms of cholesterol homeostasis, and suggest an important role for UII in modulating cholesterol regulation.
Collapse
Affiliation(s)
- Robert S Kiss
- Division of Cardiology, McGill University Health Center, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
24
|
Craig-Schapiro R, Kuhn M, Xiong C, Pickering EH, Liu J, Misko TP, Perrin RJ, Bales KR, Soares H, Fagan AM, Holtzman DM. Multiplexed immunoassay panel identifies novel CSF biomarkers for Alzheimer's disease diagnosis and prognosis. PLoS One 2011; 6:e18850. [PMID: 21526197 PMCID: PMC3079734 DOI: 10.1371/journal.pone.0018850] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 03/21/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Clinicopathological studies suggest that Alzheimer's disease (AD) pathology begins ∼10-15 years before the resulting cognitive impairment draws medical attention. Biomarkers that can detect AD pathology in its early stages and predict dementia onset would, therefore, be invaluable for patient care and efficient clinical trial design. We utilized a targeted proteomics approach to discover novel cerebrospinal fluid (CSF) biomarkers that can augment the diagnostic and prognostic accuracy of current leading CSF biomarkers (Aβ42, tau, p-tau181). METHODS AND FINDINGS Using a multiplexed Luminex platform, 190 analytes were measured in 333 CSF samples from cognitively normal (Clinical Dementia Rating [CDR] 0), very mildly demented (CDR 0.5), and mildly demented (CDR 1) individuals. Mean levels of 37 analytes (12 after Bonferroni correction) were found to differ between CDR 0 and CDR>0 groups. Receiver-operating characteristic curve analyses revealed that small combinations of a subset of these markers (cystatin C, VEGF, TRAIL-R3, PAI-1, PP, NT-proBNP, MMP-10, MIF, GRO-α, fibrinogen, FAS, eotaxin-3) enhanced the ability of the best-performing established CSF biomarker, the tau/Aβ42 ratio, to discriminate CDR>0 from CDR 0 individuals. Multiple machine learning algorithms likewise showed that the novel biomarker panels improved the diagnostic performance of the current leading biomarkers. Importantly, most of the markers that best discriminated CDR 0 from CDR>0 individuals in the more targeted ROC analyses were also identified as top predictors in the machine learning models, reconfirming their potential as biomarkers for early-stage AD. Cox proportional hazards models demonstrated that an optimal panel of markers for predicting risk of developing cognitive impairment (CDR 0 to CDR>0 conversion) consisted of calbindin, Aβ42, and age. CONCLUSIONS/SIGNIFICANCE Using a targeted proteomic screen, we identified novel candidate biomarkers that complement the best current CSF biomarkers for distinguishing very mildly/mildly demented from cognitively normal individuals. Additionally, we identified a novel biomarker (calbindin) with significant prognostic potential.
Collapse
Affiliation(s)
- Rebecca Craig-Schapiro
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Max Kuhn
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Chengjie Xiong
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Eve H. Pickering
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Jingxia Liu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Thomas P. Misko
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Richard J. Perrin
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Neuropathology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kelly R. Bales
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Holly Soares
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|