1
|
He K, Zhao Z, Zhang J, Li D, Wang S, Liu Q. Cholesterol Metabolism in Neurodegenerative Diseases. Antioxid Redox Signal 2024. [PMID: 38842175 DOI: 10.1089/ars.2024.0674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Significance: Cholesterol plays a crucial role in the brain, where it is highly concentrated and tightly regulated to support normal brain functions. It serves as a vital component of cell membranes, ensuring their integrity, and acts as a key regulator of various brain processes. Dysregulation of cholesterol metabolism in the brain has been linked to impaired brain function and the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. Recent Advances: A significant advancement has been the identification of astrocyte-derived apoliprotein E as a key regulator of de novo cholesterol biosynthesis in neurons, providing insights into how extracellular signals influence neuronal cholesterol levels. In addition, the development of antibody-based therapies, particularly for AD, presents promising opportunities for therapeutic interventions. Critical Issues: Despite significant research, the association between cholesterol and neurodegenerative diseases remains inconclusive. It is crucial to distinguish between plasma cholesterol and brain cholesterol, as these pools are relatively independent. This differentiation should be considered when evaluating statin-based treatment approaches. Furthermore, assessing not only the total cholesterol content in the brain but also its distribution among different types of brain cells is essential. Future Direction: Establishing a causal link between changes in brain/plasma cholesterol levels and the onset of brain dysfunction/neurodegenerative diseases remains a key objective. In addition, conducting cell-specific analyses of cholesterol homeostasis in various types of brain cells under pathological conditions will enhance our understanding of cholesterol metabolism in neurodegenerative diseases. Manipulating cholesterol levels to restore homeostasis may represent a novel approach for alleviating neurological symptoms.
Collapse
Affiliation(s)
- Keqiang He
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| |
Collapse
|
2
|
Shishioh N, Kiryu-Seo S, Abe-Dohmae S, Yokoyama S, Kiyama H. Expression of ATP-binding cassette transporter A1 is induced by nerve injury and its deficiency affects neurite tip morphology and elongation in cultured neurons. J Chem Neuroanat 2022; 125:102164. [PMID: 36122678 DOI: 10.1016/j.jchemneu.2022.102164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022]
Abstract
Axonal regeneration requires changes in the lipid dynamics of the axon membrane for growth and extension. Here, we examined the expression of genes associated with lipid transport after nerve injury. The expression of ATP-binding cassette transporter-A1 (ABCA1), which participates in the transport of cholesterol from the plasma membrane, was markedly upregulated in motor and sensory neurons after nerve injury. Stimulation of PC12 cells with the nerve growth factor induced neurite extension and ABCA1 expression predominantly in regions proximal to the neurite tip. To clarify the functional role of ABCA1 in neurite elongation, we examined the morphology of neurons cultured from conditionally-injured dorsal root ganglia from ABCA1-deficient mice. We found a significant increase in neurite branch formation in these neurons. In addition, the neurite tips of ABCA1-deficient neurons appeared excessively ruffled, and the direction of neurite elongation was unsteady. In contrast, the neurite tips of wild-type neurons were not excessively ruffled, and the neurites elongated rapidly in a stable directionally-oriented manner. Together, these findings suggest that ABCA1 plays an important role in regulating the membrane lipid composition of injured neurons and in axonal regeneration following nerve injury.
Collapse
Affiliation(s)
- Nobue Shishioh
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan; Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga 526-0829, Japan
| | - Sumiko Kiryu-Seo
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Sumiko Abe-Dohmae
- Food and Nutritional Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
3
|
Emerging role of HDL in brain cholesterol metabolism and neurodegenerative disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159123. [PMID: 35151900 DOI: 10.1016/j.bbalip.2022.159123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/07/2023]
Abstract
High-density lipoproteins (HDLs play a key role in cholesterol homeostasis maintenance in the central nervous system (CNS), by carrying newly synthesized cholesterol from astrocytes to neurons, to support their lipid-related physiological functions. As occurs for plasma HDLs, brain lipoproteins are assembled through the activity of membrane cholesterol transporters, undergo remodeling mediated by specific enzymes and transport proteins, and finally deliver cholesterol to neurons by a receptor-mediated internalization process. A growing number of evidences indicates a strong association between alterations of CNS cholesterol homeostasis and neurodegenerative disorders, in particular Alzheimer's disease (AD), and a possible role in this relationship may be played by defects in brain HDL metabolism. In the present review, we summarize and critically examine the current state of knowledge on major modifications of HDL and HDL-mediated brain cholesterol transport in AD, by taking into consideration the individual steps of this process. We also describe potential and encouraging HDL-based therapies that could represent new therapeutic strategies for AD treatment. Finally, we revise the main plasma and brain HDL modifications in other neurodegenerative disorders including Parkinson's disease (PD), Huntington's disease (HD), and frontotemporal dementia (FTD).
Collapse
|
4
|
Dash R, Mitra S, Ali MC, Oktaviani DF, Hannan MA, Choi SM, Moon IS. Phytosterols: Targeting Neuroinflammation in Neurodegeneration. Curr Pharm Des 2021; 27:383-401. [PMID: 32600224 DOI: 10.2174/1381612826666200628022812] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/02/2020] [Indexed: 11/22/2022]
Abstract
Plant-derived sterols, phytosterols, are well known for their cholesterol-lowering activity in serum and their anti-inflammatory activities. Recently, phytosterols have received considerable attention due to their beneficial effects on various non-communicable diseases, and recommended use as daily dietary components. The signaling pathways mediated in the brain by phytosterols have been evaluated, but little is known about their effects on neuroinflammation, and no clinical studies have been undertaken on phytosterols of interest. In this review, we discuss the beneficial roles of phytosterols, including their attenuating effects on inflammation, blood cholesterol levels, and hallmarks of the disease, and their regulatory effects on neuroinflammatory disease pathways. Despite recent advancements made in phytosterol pharmacology, some critical questions remain unanswered. Therefore, we have tried to highlight the potential of phytosterols as viable therapeutics against neuroinflammation and to direct future research with respect to clinical applications.
Collapse
Affiliation(s)
- Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Sarmistha Mitra
- Plasma Bioscience Research Center, Plasma Bio-display, Kwangwoon University, Seoul-01897, Korea
| | - Md Chayan Ali
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Diyah Fatimah Oktaviani
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Md Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Sung Min Choi
- Department of Pediatrics, Dongguk University College of Medicine, Gyeongju-38066, Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| |
Collapse
|
5
|
Santa Cruz EC, Zandonadi FDS, Fontes W, Sussulini A. A pilot study indicating the dysregulation of the complement and coagulation cascades in treated schizophrenia and bipolar disorder patients. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140657. [PMID: 33839315 DOI: 10.1016/j.bbapap.2021.140657] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/15/2022]
Abstract
A better understanding of the proteome profile after bipolar disorder (BD) and schizophrenia (SCZ) treatment, besides monitoring disease progression, may assist on the development of novel therapeutic strategies with the ability to reduce or control possible side effects. In this pilot study, proteomics analysis employing nano liquid chromatography coupled to mass spectrometry (nLC-MS) and bioinformatic tools were applied to identify differentially abundant proteins in serum of treated BD and SCZ patients. In total, 10 BD patients, 10 SCZ patients, and 14 healthy controls (HC) were included in this study. 24 serum proteins were significantly altered (p < 0.05) in BD and SCZ treated patients and, considering log2FC > 0.58, 8 proteins presented lower abundance in the BD group, while 7 proteins presented higher abundance and 2 lower abundance in SCZ group when compared against HC. Bioinformatics analysis based on these 24 proteins indicated two main altered pathways previously described in the literature; furthermore, it revealed that opposite abundances of the complement and coagulation cascades were the most significant biological processes involved in these pathologies. Moreover, we describe disease-related proteins and pathways associations suggesting the necessity of clinical follow-up improvement besides treatment, as a precaution or safety measure, along with the disease progression. Further biological validation and investigations are required to define whether there is a correlation between complement and coagulation cascade expression for BD and SCZ and cardiovascular diseases.
Collapse
Affiliation(s)
- Elisa Castañeda Santa Cruz
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil
| | - Flávia da Silva Zandonadi
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia (UnB), 70910-900 Brasilia, DF, Brazil
| | - Alessandra Sussulini
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
6
|
Chauhan MZ, Arcuri J, Park KK, Zafar MK, Fatmi R, Hackam AS, Yin Y, Benowitz L, Goldberg JL, Samarah M, Bhattacharya SK. Multi-Omic Analyses of Growth Cones at Different Developmental Stages Provides Insight into Pathways in Adult Neuroregeneration. iScience 2020; 23:100836. [PMID: 32058951 PMCID: PMC6997871 DOI: 10.1016/j.isci.2020.100836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Growth cones (GCs) are structures associated with growing neurons. GC membrane expansion, which necessitates protein-lipid interactions, is critical to axonal elongation in development and in adult neuritogenesis. We present a multi-omic analysis that integrates proteomics and lipidomics data for the identification of GC pathways, cell phenotypes, and lipid-protein interactions, with an analytic platform to facilitate the visualization of these data. We combine lipidomic data from GC and adult axonal regeneration following optic nerve crush. Our results reveal significant molecular variability in GCs across developmental ages that aligns with the upregulation and downregulation of lipid metabolic processes and correlates with distinct changes in the lipid composition of GC plasmalemma. We find that these processes also define the transition into a growth-permissive state in the adult central nervous system. The insight derived from these analyses will aid in promoting adult regeneration and functional innervation in devastating neurodegenerative diseases. Simultaneous proteomics and lipidomics analyses of developmental growth cones Combined multi-omics analyses of regenerating optic nerves and growth cones Integrating protein-protein with protein-lipid interactions in growth cones
Collapse
Affiliation(s)
- Muhammad Zain Chauhan
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jennifer Arcuri
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Program in Biomedical Sciences & Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kevin K Park
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Program in Biomedical Sciences & Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maroof Khan Zafar
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Rabeet Fatmi
- Department of Computer Science, Florida Polytechnic University, Lakeland, FL 33805, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Program in Biomedical Sciences & Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yuqin Yin
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Larry Benowitz
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jeffrey L Goldberg
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad Samarah
- Department of Computer Science, Florida Polytechnic University, Lakeland, FL 33805, USA
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Program in Biomedical Sciences & Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
7
|
Potashkin J, Huang X, Becker C, Chen H, Foltynie T, Marras C. Understanding the links between cardiovascular disease and Parkinson's disease. Mov Disord 2020; 35:55-74. [PMID: 31483535 PMCID: PMC6981000 DOI: 10.1002/mds.27836] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/16/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Studies investigating the associations between genetic or environmental factors and Parkinson's disease (PD) have uncovered a number of factors shared with cardiovascular disease, either as risk factors or manifestations of cardiovascular disease itself. Older age, male sex, and possibly type 2 diabetes are examples. On the other hand, coffee consumption and physical activity are each associated with a lower risk of both PD and cardiovascular disease. This observation raises questions about the underlying pathophysiological links between cardiovascular disease and PD. There is evidence for common mechanisms in the areas of glucose metabolism, cellular stress, lipid metabolism, and inflammation. On the other hand, smoking and total/low-density lipoprotein cholesterol appear to have opposite associations with cardiovascular disease and PD. Thus, it is uncertain whether the treatment of cardiovascular risk factors will impact on the onset or progression of PD. The available data suggest that a nuanced approach is necessary to manage risk factors such as cholesterol levels once the associations are better understood. Ultimately, the choice of therapy may be tailored to a patient's comorbidity profile. This review presents the epidemiological evidence for both concordant and discordant associations between cardiovascular disease and PD, discusses the cellular and metabolic processes that may underlie these links, and explores the implications this has for patient care and future research. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Judy Potashkin
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Xuemei Huang
- Translational Brain Research Center and Department of Neurology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Claudia Becker
- Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
| | - Thomas Foltynie
- Department of Clinical & Movement Neurosciences, University College London Institute of Neurology, Queen Square, London, United Kingdom
| | - Connie Marras
- The Edmond J Safra Program in Parkinson's Research, Toronto Western Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
8
|
Arbo B, Ribeiro M, Garcia-Segura L. Development of new treatments for Alzheimer's disease based on the modulation of translocator protein (TSPO). Ageing Res Rev 2019; 54:100943. [PMID: 31430564 DOI: 10.1016/j.arr.2019.100943] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/07/2019] [Accepted: 08/15/2019] [Indexed: 12/27/2022]
Abstract
The increase in life expectancy of the world population is associated with a higher prevalence of neurodegenerative diseases. Alzheimer's Disease (AD) is the most common neurodegenerative disease, affecting currently 43 million people over the world. To date, most of the pharmacological interventions in AD are intended for the alleviation of some of its symptoms, and there are no effective treatments to inhibit the progression of the disease. Translocator protein (TSPO) is present in contact points between the outer and the inner mitochondrial membranes and is involved in the control of steroidogenesis, inflammation and apoptosis. In the last decade, studies have shown that TSPO ligands present neuroprotective effects in different experimental models of AD, both in vitro and in vivo. The aim of this review is to analyze the data provided by these studies and to discuss if TSPO could be a viable therapeutic target for the development of new treatments for AD.
Collapse
|
9
|
Chen X, Miller NM, Afghah Z, Geiger JD. Development of AD-Like Pathology in Skeletal Muscle. JOURNAL OF PARKINSON'S DISEASE AND ALZHEIMER'S DISEASE 2019; 6:10.13188/2376-922x.1000028. [PMID: 32190732 PMCID: PMC7079679 DOI: 10.13188/2376-922x.1000028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Effective therapeutic strategy against Alzheimer's disease (AD) requires early detection of AD; however, clinical diagnosis of Alzheimer's disease (AD) is not precise and a definitive diagnosis of AD is only possible via postmortem examination for AD pathological hallmarks including senile plaques composed of Aβ and neuro fibrillary tangles composed of phosphorylated tau. Although a variety of biomarker has been developed and used in clinical setting, none of them robustly predicts subsequent clinical course of AD. Thus, it is essential to identify new biomarkers that may facilitate the diagnosis of early stages of AD, prediction of subsequent clinical course, and development of new therapeutic strategies. Given that pathological hallmarks of AD including Aβaccumulation and the presence of phosphorylated tau are also detected in peripheral tissues, AD is considered a systemic disease. Without the protection of blood-brain barrier, systemic factors can affect peripheral tissues much earlier than neurons in brain. Here, we will discuss the development of AD-like pathology in skeletal muscle and the potential use of skeletal muscle biopsy (examination for Aβaccumulation and phosphorylated tau) as a biomarker for AD.
Collapse
Affiliation(s)
- X Chen
- Department of Biomedical Sciences, University of North Dakota, USA
| | - NM Miller
- Department of Biomedical Sciences, University of North Dakota, USA
| | - Z Afghah
- Department of Biomedical Sciences, University of North Dakota, USA
| | - JD Geiger
- Department of Biomedical Sciences, University of North Dakota, USA
| |
Collapse
|
10
|
Zhang L, Wang X, Wang M, Sterling NW, Du G, Lewis MM, Yao T, Mailman RB, Li R, Huang X. Circulating Cholesterol Levels May Link to the Factors Influencing Parkinson's Risk. Front Neurol 2017; 8:501. [PMID: 29021777 PMCID: PMC5624032 DOI: 10.3389/fneur.2017.00501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/07/2017] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVES A growing literature suggests that circulating cholesterol levels have been associated with Parkinson's disease (PD). In this study, we investigated a possible causal basis for the cholesterol-PD link. METHODS Fasting plasma cholesterol levels were obtained from 91 PD and 70 age- and gender-matched controls from an NINDS PD Biomarkers Program cohort at the Pennsylvania State University College of Medicine. Based on the literature, genetic polymorphisms in selected cholesterol management genes (APOE, LDLR, LRP1, and LRPAP1) were chosen as confounding variables because they may influence both cholesterol levels and PD risk. First, the marginal structure model was applied, where the associations of total- and LDL-cholesterol levels with genetic polymorphisms, statin usage, and smoking history were estimated using linear regression. Then, potential causal influences of total- and LDL-cholesterol on PD occurrence were investigated using a generalized propensity score approach in the second step. RESULTS Both statins (p < 0.001) and LRP1 (p < 0.03) influenced total- and LDL-cholesterol levels. There also was a trend for APOE to affect total- and LDL-cholesterol (p = 0.08 for both), and for LRPAR1 to affect LDL-cholesterol (p = 0.05). Conversely, LDLR did not influence plasma cholesterol levels (p > 0.19). Based on propensity score methods, lower total- and LDL-cholesterol were significantly linked to PD (p < 0.001 and p = 0.04, respectively). CONCLUSION The current study suggests that circulating total- and LDL-cholesterol levels potentially may be linked to the factor(s) influencing PD risk. Further studies to validate these results would impact our understanding of the role of cholesterol as a risk factor in PD, and its relationship to recent public health controversies.
Collapse
Affiliation(s)
- Lijun Zhang
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Xue Wang
- Department of Industrial and Manufacturing Engineering, Pennsylvania State University, University Park, PA, United States
| | - Ming Wang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Nick W. Sterling
- Department of Neurology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Pharmacology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Tao Yao
- Department of Industrial and Manufacturing Engineering, Pennsylvania State University, University Park, PA, United States
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Pharmacology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Runze Li
- Department of Statistics, Pennsylvania State University, University Park, PA, United States
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Pharmacology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Radiology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Neurosurgery, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Kinesiology, Pennsylvania State University College of Medicine-Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
11
|
Ubaida-Mohien C, Lamberty B, Dickens AM, Mielke MM, Marcotte T, Sacktor N, Grant I, Letendre S, Franklin D, Cibrowski P, Tharakan R, McArthur JC, Fox H, Haughey NJ. Modifications in acute phase and complement systems predict shifts in cognitive status of HIV-infected patients. AIDS 2017; 31:1365-1378. [PMID: 28574961 PMCID: PMC5501712 DOI: 10.1097/qad.0000000000001503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The prevalence of HIV-associated neurocognitive disorders (HAND) has not changed considerably in the last two decades. Potent antiretroviral therapy has shifted the severity of HAND to milder phenotypes, but excess morbidity and mortality continue to be associated with HAND. Changes in numerous markers of immune function, inflammation, and cellular stress have been repeatedly associated with HAND, but the underlying systems that drive these changes have not been identified. METHOD In this study, we used systems informatics to interrogate the cerebrospinal fluid proteomic content of longitudinal samples obtained from HIV-infected adults with stably unimpaired, stably impaired, worsening, or improving neurocognitive performance. RESULTS AND CONCLUSION The patterns of change in cerebrospinal fluid protein content implicated the induction of acute phase and complement systems as important regulators of neurocognitive status. Worsening neurocognitive performance was preceded by induction of acute phase and complement systems, whereas improving neurocognitive performance was preceded by a downregulation of these systems.
Collapse
Affiliation(s)
- Ceereena Ubaida-Mohien
- Intramural Research Program, National Institute on Aging, Baltimore,
Maryland
- The Johns Hopkins University School of Medicine, Department of
Neurology, Baltimore, MD
| | - Benjamin Lamberty
- The University of Nebraska Medical Center, Department of
Pharmacology and Experimental Neuroscience, Omaha, NE
| | - Alex M. Dickens
- The Johns Hopkins University School of Medicine, Department of
Neurology, Baltimore, MD
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research and
Department of Neurology College of Medicine, Mayo Clinic, Rochester, MN
| | - Thomas Marcotte
- HIV Neurobehavioral Research Program and Department of Psychiatry,
School of Medicine, University of California, San Diego, La Jolla, CA
| | - Ned Sacktor
- The Johns Hopkins University School of Medicine, Department of
Neurology, Baltimore, MD
| | - Igor Grant
- HIV Neurobehavioral Research Program and Department of Psychiatry,
School of Medicine, University of California, San Diego, La Jolla, CA
| | - Scott Letendre
- HIV Neurobehavioral Research Program and Department of Psychiatry,
School of Medicine, University of California, San Diego, La Jolla, CA
| | - D Franklin
- HIV Neurobehavioral Research Program and Department of Psychiatry,
School of Medicine, University of California, San Diego, La Jolla, CA
| | - Pawel Cibrowski
- The University of Nebraska Medical Center, Department of
Pharmacology and Experimental Neuroscience, Omaha, NE
| | - Ravi Tharakan
- The Johns Hopkins University School of Medicine, Department of
Psychiatry, Baltimore, MD
| | - Justin C. McArthur
- The Johns Hopkins University School of Medicine, Department of
Neurology, Baltimore, MD
| | - Howard Fox
- The University of Nebraska Medical Center, Department of
Pharmacology and Experimental Neuroscience, Omaha, NE
| | - Norman J. Haughey
- The Johns Hopkins University School of Medicine, Department of
Neurology, Baltimore, MD
- The Johns Hopkins University School of Medicine, Department of
Psychiatry, Baltimore, MD
| |
Collapse
|
12
|
El Azzouzi K, Wiesner C, Linder S. Metalloproteinase MT1-MMP islets act as memory devices for podosome reemergence. J Cell Biol 2016; 213:109-25. [PMID: 27069022 PMCID: PMC4828691 DOI: 10.1083/jcb.201510043] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 03/02/2016] [Indexed: 12/11/2022] Open
Abstract
The authors find that matrix metalloproteinase MT1-MMP is enriched at the plasma membrane of macrophage podosomes, where it persists beyond podosome lifetime and, through binding to the subcortical actin cytoskeleton, forms subcellular signposts that facilitate podosome reformation. Podosomes are dynamic cell adhesions that are also sites of extracellular matrix degradation, through recruitment of matrix-lytic enzymes, particularly of matrix metalloproteinases. Using total internal reflection fluorescence microscopy, we show that the membrane-bound metalloproteinase MT1-MMP is enriched not only at podosomes but also at distinct “islets” embedded in the plasma membrane of primary human macrophages. MT1-MMP islets become apparent upon podosome dissolution and persist beyond podosome lifetime. Importantly, the majority of MT1-MMP islets are reused as sites of podosome reemergence. siRNA-mediated knockdown and recomplementation analyses show that islet formation is based on the cytoplasmic tail of MT1-MMP and its ability to bind the subcortical actin cytoskeleton. Collectively, our data reveal a previously unrecognized phase in the podosome life cycle and identify a structural function of MT1-MMP that is independent of its proteolytic activity. MT1-MMP islets thus act as cellular memory devices that enable efficient and localized reformation of podosomes, ensuring coordinated matrix degradation and invasion.
Collapse
Affiliation(s)
- Karim El Azzouzi
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, 20246 Hamburg, Germany
| | - Christiane Wiesner
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, 20246 Hamburg, Germany
| | - Stefan Linder
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW This article evaluates recent experimental and human evidence regarding the involvement of lipids, lipoproteins, and apolipoproteins in neurodegenerative diseases, and reviews the current literature of the effects of cholesterol-lowering treatment on cognition. RECENT FINDINGS Plasma levels of traditional lipids and lipoproteins are not consistently associated with risk of dementia even though low plasma levels of apolipoprotein E, through unknown mechanisms, robustly predict future dementia. Experimental evidence suggests neuroprotective roles of several brain and cerebrospinal fluid apolipoproteins. Whether plasma levels of apolipoprotein E, or any other apolipoprotein with possible central nervous system and/or blood-brain barrier functions (apolipoproteins J, A-I, A-II, A-IV, D, C-I, and C-III) may become accessible biomarker components that improve risk prediction for dementia together with genetic risk variants and cardiovascular risk factors remains to be determined. SUMMARY Apolipoproteins with well established functions in peripheral lipid metabolism may play important roles for brain vascular health and Alzheimer's disease pathophysiology. Experimental work on lipids, lipoproteins, and apolipoproteins in the central nervous system together with robust prospective human studies will help to substantiate the drug target potential of these lipid components.
Collapse
Affiliation(s)
- Cheryl L Wellington
- aDepartment of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada bDepartment of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospitals cFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
14
|
Sterling NW, Lichtenstein M, Lee EY, Lewis MM, Evans A, Eslinger PJ, Du G, Gao X, Chen H, Kong L, Huang X. Higher Plasma LDL-Cholesterol is Associated with Preserved Executive and Fine Motor Functions in Parkinson's Disease. Aging Dis 2016; 7:237-45. [PMID: 27330838 PMCID: PMC4898920 DOI: 10.14336/ad.2015.1030] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/30/2015] [Indexed: 11/26/2022] Open
Abstract
Plasma low density lipoprotein (LDL) cholesterol has been associated both with risk of Parkinson's disease (PD) and with age-related changes in cognitive function. This prospective study examined the relationship between baseline plasma LDL-cholesterol and cognitive changes in PD and matched Controls. Fasting plasma LDL-cholesterol levels were obtained at baseline from 64 non-demented PD subjects (62.7 ± 7.9 y) and 64 Controls (61.3 ± 6.8 y). Subjects underwent comprehensive neuropsychological testing at baseline, 18-, and 36-months. Linear mixed-effects modeling was used to assess the relationships between baseline LDL-cholesterol levels and longitudinal cognitive changes. At baseline, PD patients had lower scores of fine motor (p<0.0001), executive set shifting (p=0.018), and mental processing speed (p=0.049) compared to Controls. Longitudinally, Controls demonstrated improved fine motor and memory test scores (p=0.044, and p=0.003), whereas PD patients demonstrated significantly accelerated loss in fine motor skill (p=0.002) compared to Controls. Within the PD group, however, higher LDL-cholesterol levels were associated with improved executive set shifting (β=0.003, p<0.001) and fine motor scores (β=0.002, p=0.030) over time. These associations were absent in Controls (p>0.7). The cholesterol - executive set shifting association differed significantly between PDs and Controls (interaction p=0.005), whereas the cholesterol - fine motor association difference did not reach significance (interaction, p=0.104). In summary, higher plasma LDL-cholesterol levels were associated with better executive function and fine motor performance over time in PD, both of which may reflect an effect on nigrostriatal mediation. Confirmation of these results and elucidation of involved mechanisms are warranted, and might lead to feasible therapeutic strategies.
Collapse
Affiliation(s)
- Nicholas W. Sterling
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Maya Lichtenstein
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Eun-Young Lee
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Mechelle M. Lewis
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Alicia Evans
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Paul J. Eslinger
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Guangwei Du
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Xiang Gao
- Department of Nutritional Sciences, the Pennsylvania State University, University Park, PA 16802, USA.
| | - Honglei Chen
- Epidemiology Branch/Aging & Neuroepidemiology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Lan Kong
- Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| | - Xuemei Huang
- Departments of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
- Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA.
| |
Collapse
|
15
|
Glodzik L, Rusinek H, Kamer A, Pirraglia E, Tsui W, Mosconi L, Li Y, McHugh P, Murray J, Williams S, Osorio RS, Randall C, Butler T, Deshpande A, Vallabhajolusa S, de Leon M. Effects of vascular risk factors, statins, and antihypertensive drugs on PiB deposition in cognitively normal subjects. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2016; 2:95-104. [PMID: 27239540 PMCID: PMC4879519 DOI: 10.1016/j.dadm.2016.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Hypertension, hypercholesterolemia, and obesity increase the risk of dementia. Although their detection is commonly followed by an introduction of treatment, little is known about how medications frequently used to treat vascular risk affect amyloid deposition. METHODS A cross-sectional study of 156 subjects who underwent positron emission tomography with PiB. Using linear regression, we tested whether blood pressure, cholesterol, overweight/obese status, angiotensin receptor blockers (ARBs), beta-blockers, diuretics, angiotensin converting enzyme inhibitors, and statins predicted amyloid deposition. RESULTS The use of ARBs (β = -.15, P = .044) and diuretics (β = -.20, P = .006) predicted less amyloid accumulation; older age (β = .29, P < .001) and statins (β = .23, P = .004) were related to greater amyloid deposition. Overweight and/or obese women had more cortical amyloid than their peers. DISCUSSION Prospective studies should confirm effects of drugs and increased body weight on amyloid accumulation and establish whether they translate into measurable clinical outcomes. Women may be more susceptible to harmful effects of obesity.
Collapse
Affiliation(s)
- Lidia Glodzik
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
- Department of Radiology, NYU School of Medicine, New York, USA
- Corresponding author. Tel.: +1 212 263 5698; Fax: +1 212 263 3270.
| | - Henry Rusinek
- Department of Radiology, NYU School of Medicine, New York, USA
| | - Angela Kamer
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, New York, USA
| | - Elizabeth Pirraglia
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Wai Tsui
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Lisa Mosconi
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Yi Li
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Pauline McHugh
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - John Murray
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | | | - Ricardo S. Osorio
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Catherine Randall
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Tracy Butler
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Anup Deshpande
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | | | - Mony de Leon
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| |
Collapse
|
16
|
Yan X, Ma L, Hovakimyan M, Lukas J, Wree A, Frank M, Guthoff R, Rolfs A, Witt M, Luo J. Defects in the retina of Niemann-pick type C 1 mutant mice. BMC Neurosci 2014; 15:126. [PMID: 25472750 PMCID: PMC4267119 DOI: 10.1186/s12868-014-0126-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/12/2014] [Indexed: 11/24/2022] Open
Abstract
Background Niemann-Pick type C1 (NPC1) disease is an inherited lysosomal storage disease caused by mutation of the Npc1 gene, resulting in a progressive accumulation of unesterified cholesterol and glycolipids in lysosomes of multiple tissues and leading to neurodegeneration and other disease. In Npc1 mutant mice, retinal degeneration including impaired visual function, lipofuscin accumulation in the pigment epithelium and ganglion cells as well as photoreceptor defects has been found. However, the pathologies of other individual cell types of the retina in Npc1 mutant mice are still not fully clear. We hypothesized that horizontal cells, amacrine cells, bipolar cells and glial cells are also affected in the retina of Npc1 mutant mice. Results Immunohistochemistry and electron microscopy were used to investigate pathologies of ganglion cells, horizontal cells, amacrine cells, bipolar cells, and optic nerves as well as altered activity of glial cells in Npc1 mutant mice. Electron microscopy reveals that electron-dense inclusions are generally accumulated in ganglion cells, bipolar cells, Müller cells, and in the optic nerve. Furthermore, abnormal arborisation and ectopic processes of horizontal and amacrine cells as well as defective bipolar cells are observed by immunohistochemistry for specific cellular markers. Furthermore, hyperactivity of glial cells, including astrocytes, microglial cells, and Müller cells, is also revealed. Conclusions Our data extend previous findings to show multiple defects in the retina of Npc1 mutant mice, suggesting an important role of Npc1 protein in the normal function of the retina. Electronic supplementary material The online version of this article (doi:10.1186/s12868-014-0126-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Yan
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Strasse 20, D-18147, Rostock, Germany.
| | - Lucy Ma
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Strasse 20, D-18147, Rostock, Germany.
| | - Marina Hovakimyan
- Institute for Biomedical Engineering, Rostock University Medical Center, F.-Barnewitz Strasse 4, D-18119, Rostock, Germany.
| | - Jan Lukas
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Strasse 20, D-18147, Rostock, Germany.
| | - Andreas Wree
- Department of Anatomy, Rostock University Medical Center, Gertrudenstrsse 9, D-18055, Rostock, Germany.
| | - Marcus Frank
- Electron Microscopy Center, Rostock University Medical Center, Strempelstr. 14, D-18057, Rostock, Germany.
| | - Rudolf Guthoff
- Department of Ophthalmology, Rostock University Medical Center, Doberaner Strasse 140, D-18057, Rostock, Germany.
| | - Arndt Rolfs
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Strasse 20, D-18147, Rostock, Germany.
| | - Martin Witt
- Department of Anatomy, Rostock University Medical Center, Gertrudenstrsse 9, D-18055, Rostock, Germany.
| | - Jiankai Luo
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Strasse 20, D-18147, Rostock, Germany.
| |
Collapse
|
17
|
Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, Rakugi H, Morishita R. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Front Aging Neurosci 2014; 6:71. [PMID: 24795626 PMCID: PMC4005936 DOI: 10.3389/fnagi.2014.00071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/30/2014] [Indexed: 12/28/2022] Open
Abstract
The benefits of statins, commonly prescribed for hypercholesterolemia, in treating Alzheimer's disease (AD) have not yet been fully established. A recent randomized clinical trial did not show any therapeutic effects of two statins on cognitive function in AD. Interestingly, however, the results of the Rotterdam study, one of the largest prospective cohort studies, showed reduced risk of AD in statin users. Based on the current understanding of statin actions and AD pathogenesis, it is still worth exploring whether statins can prevent AD when administered decades before the onset of AD or from midlife. This review discusses the possible beneficial effects of statins, drawn from previous clinical observations, pathogenic mechanisms, which include β-amyloid (Aβ) and tau metabolism, genetic and non-genetic risk factors (apolipoprotein E, cholesterol, sex, hypertension, and diabetes), and other clinical features (vascular dysfunction and oxidative and inflammatory stress) of AD. These findings suggest that administration of statins in midlife might prevent AD in late life by modifying genetic and non-genetic risk factors for AD. It should be clarified whether statins inhibit Aβ accumulation, tau pathological features, and brain atrophy in humans. To answer this question, a randomized controlled study using amyloid positron emission tomography (PET), tau-PET, and magnetic resonance imaging would be useful. This clinical evaluation could help us to overcome this devastating disease.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Hitomi Kurinami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Toshimitsu Hamasaki
- Department of Biomedical Statistics, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Amarnath Chatterjee
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| |
Collapse
|
18
|
Chen X, Hui L, Soliman ML, Geiger JD. Altered Cholesterol Intracellular Trafficking and the Development of Pathological Hallmarks of Sporadic AD. ACTA ACUST UNITED AC 2014; 1. [PMID: 25621310 DOI: 10.13188/2376-922x.1000002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Compared to the rare familial early onset Alzheimer's disease (AD) that results from gene mutations in AbPP and presenilin-1, the pathogenesis of sporadic AD is much more complex and is believed to result from complex interactions between nutritional, environmental, epigenetic and genetic factors. Among those factors, the presence APOE4 is still the single strongest genetic risk factor for sporadic AD. However, the exact underlying mechanism whereby apoE4 contributes to the pathogenesis of sporadic AD remains unclear. Here, we discuss how altered cholesterol intracellular trafficking as a result of apoE4 might contribute to the development of pathological hallmarks of AD including brain deposition of amyloid beta (Ab), neurofibrillary tangles, and synaptic dysfunction.
Collapse
|
19
|
Chen X, Hui L, Geiger JD. Role of LDL cholesterol and endolysosomes in amyloidogenesis and Alzheimer's disease. ACTA ACUST UNITED AC 2014; 5. [PMID: 26413387 DOI: 10.4172/2155-9562.1000236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The pathogenesis of late-onset sporadic Alzheimer's disease (AD) is believed to result from complex interactions between nutritional, environmental, epigenetic and genetic factors. Among those factors, altered circulating cholesterol homeostasis, independent of the APOE genotype, continues to be implicated in brain deposition of amyloid beta protein (Aβ) and the pathogenesis of AD. It is believed that trafficking of amyloid beta precursor protein (AβPP) into endolysosomes appears to play a critical role in determining amyloidogenic processing of AβPP because this is precisely where two enzymes critically important in AβPP metabolism are located; beta amyloid converting enzyme (BACE-1) and gamma secretase enzyme. We have shown that elevated levels of LDL cholesterol promote AβPP internalization, disturb neuronal endolysosome structure and function, and increase Aβ accumulation in neuronal endolysosomes. Here, we will further discuss the linkage between elevated levels of LDL cholesterol and AD pathogenesis, and explore the underlying mechanisms whereby elevated levels of plasma LDL cholesterol promote amyloidogenesis.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Liang Hui
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|
20
|
|
21
|
A suppressor screen in Mecp2 mutant mice implicates cholesterol metabolism in Rett syndrome. Nat Genet 2013; 45:1013-20. [PMID: 23892605 PMCID: PMC3837522 DOI: 10.1038/ng.2714] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/24/2013] [Indexed: 12/13/2022]
Abstract
Mutations in methyl CpG binding protein 2 (MECP2) cause Rett Syndrome, the most severe autism spectrum disorder. Re-expressing Mecp2 in symptomatic Mecp2 null mice dramatically improves function and longevity, providing hope that therapeutic intervention is possible in humans. To identify pathways in disease pathology for therapeutic intervention, a dominant ENU mutagenesis suppressor screen was carried out in Mecp2 null mice. Five suppressors that ameliorate symptoms of Mecp2 loss were isolated. Here we show that a stop codon mutation in squalene epoxidase (Sqle), a rate-limiting enzyme in cholesterol biosynthesis underlies suppression in one line. Subsequently, we show that lipid metabolism is perturbed in the brain and liver of Mecp2 null males. Consistently, statin drugs improve systemic perturbations of lipid metabolism, alleviate motor symptoms and confer increased longevity in Mecp2 mutant mice. The genetic screen therefore points to cholesterol homeostasis as a potential target for the treatment of Rett patients.
Collapse
|
22
|
Bergamin N, Dardis A, Beltrami A, Cesselli D, Rigo S, Zampieri S, Domenis R, Bembi B, Beltrami CA. A human neuronal model of Niemann Pick C disease developed from stem cells isolated from patient's skin. Orphanet J Rare Dis 2013; 8:34. [PMID: 23433359 PMCID: PMC3648447 DOI: 10.1186/1750-1172-8-34] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 02/14/2013] [Indexed: 11/10/2022] Open
Abstract
Background Niemann Pick C (NPC) disease is a neurovisceral lysosomal storage disorder due to mutations in NPC1 or NPC2 genes, characterized by the accumulation of endocytosed unesterified cholesterol, gangliosides and other lipids within the lysosomes/late endosomes. Even if the neurodegeneration is the main feature of the disease, the analysis of the molecular pathways linking the lipid accumulation and cellular damage in the brain has been challenging due to the limited availability of human neuronal models. Objective The aim of this study was to develop a human neuronal model of NPC disease by inducing neuronal differentiation of multipotent adult stem cells (MASC) isolated from NPC patients. Methods Stem cells were isolated from 3 NPC patients and 3 controls both from skin biopsies and previously established skin fibroblast cultures. Cells were induced to differentiate along a neuronal fate adapting methods previously described by Beltrami et al, 2007. The surface immunophenotype of stem cells was analyzed by FACS. Stem cell and neuronal markers expression were evaluated by immunofluorescence. Intracellular accumulation of cholesterol and gangliosides were assessed by filipin staining and immunofluorescence, respectively. A morphometric analysis was performed using a Neurite outgrowth image program. Results After 3 passages in selective medium, MASC isolated either from skin biopsies or previously established skin fibroblast cultures displayed an antigenic pattern characteristic of mesenchymal stem cells and expressed the stem cell markers Oct-4, Nanog, Sox-2 and nestin. A massive lysosomal accumulation of cholesterol was observed only in cells isolated from NPC patients. After the induction of neural differentiation, remarkable morphologic changes were observed and cells became positive to markers of the neuronal lineage NeuN and MAP2. Differentiated cells from NPC patients displayed characteristic features of NPC disease, they showed intracellular accumulation of unesterified cholesterol and GM2 ganglioside and presented morphological differences with respect to cells derived from healthy donors. In conclusion, we generated a human neuronal model of NPC disease through the induction of differentiation of stem cells obtained from patient’s easily accessible sources. The strategy described here may be applied to easily generate human neuronal models of other neurodegenerative diseases.
Collapse
Affiliation(s)
- Natascha Bergamin
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chen J, Zhang X, Kusumo H, Costa LG, Guizzetti M. Cholesterol efflux is differentially regulated in neurons and astrocytes: implications for brain cholesterol homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:263-75. [PMID: 23010475 DOI: 10.1016/j.bbalip.2012.09.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 09/05/2012] [Accepted: 09/14/2012] [Indexed: 11/15/2022]
Abstract
Disruption of cholesterol homeostasis in the central nervous system (CNS) has been associated with neurological, neurodegenerative, and neurodevelopmental disorders. The CNS is a closed system with regard to cholesterol homeostasis, as cholesterol-delivering lipoproteins from the periphery cannot pass the blood-brain-barrier and enter the brain. Different cell types in the brain have different functions in the regulation of cholesterol homeostasis, with astrocytes producing and releasing apolipoprotein E and lipoproteins, and neurons metabolizing cholesterol to 24(S)-hydroxycholesterol. We present evidence that astrocytes and neurons adopt different mechanisms also in regulating cholesterol efflux. We found that in astrocytes cholesterol efflux is induced by both lipid-free apolipoproteins and lipoproteins, while cholesterol removal from neurons is triggered only by lipoproteins. The main pathway by which apolipoproteins induce cholesterol efflux is through ABCA1. By upregulating ABCA1 levels and by inhibiting its activity and silencing its expression, we show that ABCA1 is involved in cholesterol efflux from astrocytes but not from neurons. Furthermore, our results suggest that ABCG1 is involved in cholesterol efflux to apolipoproteins and lipoproteins from astrocytes but not from neurons, while ABCG4, whose expression is much higher in neurons than astrocytes, is involved in cholesterol efflux from neurons but not astrocytes. These results indicate that different mechanisms regulate cholesterol efflux from neurons and astrocytes, reflecting the different roles that these cell types play in brain cholesterol homeostasis. These results are important in understanding cellular targets of therapeutic drugs under development for the treatments of conditions associated with altered cholesterol homeostasis in the CNS.
Collapse
Affiliation(s)
- Jing Chen
- Department of Environmental and Occupational Health Sciences, University of Washington, 4225 Roosevelt way NE, Seattle, WA 98105, USA
| | | | | | | | | |
Collapse
|
24
|
Endolysosome involvement in LDL cholesterol-induced Alzheimer's disease-like pathology in primary cultured neurons. Life Sci 2012; 91:1159-68. [PMID: 22580286 DOI: 10.1016/j.lfs.2012.04.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/19/2012] [Accepted: 04/24/2012] [Indexed: 12/26/2022]
Abstract
AIMS Elevated levels of circulating cholesterol are extrinsic factors contributing to the pathogenesis of sporadic Alzheimer's disease (AD). We showed previously that rabbits fed a cholesterol-enriched diet exhibited blood-brain barrier (BBB) dysfunction, increased accumulation of apolipoprotein B (ApoB) in brain neurons, and endolysosomes in brain had disturbed structures and functions. These effects were linked to increased amyloid beta (Aβ) production, increased tau-pathology, and disrupted synaptic integrity. Because pathological changes to endolysosomes represent a very early event in sporadic AD, we determined here the extent to which ApoB-containing LDL cholesterol altered the structure and function of endolysosomes and contributed to the development of AD-like pathology in primary cultured neurons. MAIN METHODS Cholesterol distribution and endolysosome morphology were determined histologically. Endolysosome pH was measured ratio-metrically with LysoSensor dye. Endolysosome enzyme activity was measured for acid phosphatase, cathepsins B and D, and beta-site APP cleaving enzyme 1 (BACE-1). AD-like pathologies, including increased production of Aβ, increased tau-pathology, and disrupted synaptic integrity were determined using ELISA, immunoblotting, and immunostaining techniques. KEY FINDINGS Treatment of neurons with ApoB-containing LDL cholesterol increased endolysosome accumulation of cholesterol, enlarged endolysosomes, and elevated endolysosome pH. In addition, ApoB-containing LDL cholesterol increased endolysosome accumulation of BACE-1, enhanced BACE-1 activity, increased Aβ levels, increased levels of phosphorylated tau, and decreased levels of synaptophysin. SIGNIFICANCE Our findings suggest strongly that alterations in the structure and function of endolysosomes play a key role in the exhibition of pathological features of AD that result from neuronal exposure to ApoB-containing LDL cholesterol.
Collapse
|
25
|
Abstract
The long-term effects of a diet rich in saturated fat and cholesterol on the hippocampus were evaluated in this study. It has previously been shown that this type of diet is detrimental to health, particularly affecting peripheral organs such as the heart and liver. However, effects on the brain have not been fully evaluated. This study focused on the hippocampus, a brain region instrumental for learning and memory and vulnerable to ischemic damage. Reduced blood-brain barrier (BBB) integrity and increased microgliosis were observed in the hippocampus of rats fed a high-saturated-fat and cholesterol (HFHC) diet for 6 months. Interestingly, an increase in hippocampal protein levels of occludin, a tight junction protein, was found in HFHC-treated rats as well. Further investigation revealed decreased expression of the occludin protein in blood vessels and increased expression in the dentate gyrus hilar neurons and mossy fibers of the hippocampal cornus ammonis 3 in HFHC-treated rats. Our results show alterations in BBB integrity and expression of tight junction proteins after long-term exposure to HFHC diet in rats. These findings may suggest a biologic mechanism for previously observed behavioral deficits occurring in rats fed this diet.
Collapse
|
26
|
Ordonez MP, Roberts EA, Kidwell CU, Yuan SH, Plaisted WC, Goldstein LSB. Disruption and therapeutic rescue of autophagy in a human neuronal model of Niemann Pick type C1. Hum Mol Genet 2012; 21:2651-62. [PMID: 22437840 DOI: 10.1093/hmg/dds090] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An unresolved issue about many neurodegenerative diseases is why neurons are particularly sensitive to defects in ubiquitous cellular processes. One example is Niemann Pick type C1, caused by defects in cholesterol trafficking in all cells, but where neurons are preferentially damaged. Understanding this selective failure is limited by the difficulty in obtaining live human neurons from affected patients. To solve this problem, we generated neurons with decreased function of NPC1 from human embryonic stem cells and used them to test the hypothesis that defective cholesterol handling leads to enhanced pathological phenotypes in neurons. We found that human NPC1 neurons have strong spontaneous activation of autophagy, and, contrary to previous reports in patient fibroblasts, a block of autophagic progression leading to defective mitochondrial clearance. Mitochondrial fragmentation is an exceptionally severe phenotype in NPC1 neurons compared with fibroblasts, causing abnormal accumulation of mitochondrial proteins. Contrary to expectation, these abnormal phenotypes were rescued by treatment with the autophagy inhibitor 3-methyladenine and by treatment with the potential therapeutic cyclodextrin, which mobilizes cholesterol from the lysosomal compartment. Our findings suggest that neurons are especially sensitive to lysosomal cholesterol accumulation because of autophagy disruption and accumulation of fragmented mitochondria, thus defining a new route to effective drug development for NPC1 disease.
Collapse
Affiliation(s)
- M Paulina Ordonez
- Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
27
|
Assembly of a new growth cone after axotomy: the precursor to axon regeneration. Nat Rev Neurosci 2012; 13:183-93. [PMID: 22334213 DOI: 10.1038/nrn3176] [Citation(s) in RCA: 327] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The assembly of a new growth cone is a prerequisite for axon regeneration after injury. Creation of a new growth cone involves multiple processes, including calcium signalling, restructuring of the cytoskeleton, transport of materials, local translation of messenger RNAs and the insertion of new membrane and cell surface molecules. In axons that have an intrinsic ability to regenerate, these processes are executed in a timely fashion. However, in axons that lack regenerative capacity, such as those of the mammalian CNS, several of the steps that are required for regeneration fail, and these axons do not begin the growth process. Identification of the points of failure can suggest targets for promoting regeneration.
Collapse
|
28
|
Hayashi H. Lipid metabolism and glial lipoproteins in the central nervous system. Biol Pharm Bull 2011; 34:453-61. [PMID: 21467629 DOI: 10.1248/bpb.34.453] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lipoproteins in the central nervous system (CNS) are not incorporated from the blood but are formed mainly by glial cells within the CNS. In addition, cholesterol in the CNS is synthesized endogenously because the blood-brain barrier segregates the CNS from the peripheral circulation. Apolipoprotein (apo) E is a major apo in the CNS. In normal condition, apo E is secreted from glia, mainly from astrocytes, and forms cholesterol-rich lipoproteins by ATP-binding cassette transporters. Subsequently, apo E-containing glial lipoproteins supply cholesterol and other components to neurons via a receptor-mediated process. Recent findings demonstrated that receptors of the low density lipoprotein (LDL) receptor family not only internalize lipoproteins into the cells but also, like signaling receptors, transduce signals upon binding the ligands. In this review, the regulation of lipid homeostasis will be discussed as well as roles of lipoproteins and functions of receptors of LDL receptor family in the CNS. Furthermore, the relation between lipid metabolism and Alzheimer's disease (AD) is discussed.
Collapse
Affiliation(s)
- Hideki Hayashi
- Priority Organization for Innovation and Excellence, Kumamoto University, Honjo, Japan.
| |
Collapse
|
29
|
Freeman LR, Haley-Zitlin V, Stevens C, Granholm AC. Diet-induced effects on neuronal and glial elements in the middle-aged rat hippocampus. Nutr Neurosci 2011; 14:32-44. [PMID: 21535919 DOI: 10.1179/174313211x12966635733358] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Consumption of a high-fat and/or high-cholesterol diet can have detrimental effects on the brain. In the present study, dietary treatment with saturated fats, trans fats, or cholesterol to middle-aged Fischer 344 rats resulted in alterations to serum triglyceride and cholesterol levels, organ weights, and hippocampal morphology. Previously, we demonstrated that a 10% hydrogenated coconut oil and 2% cholesterol diet resulted in worse performance on the 12-day water radial arm maze, increased cholesterol and triglyceride levels, and decreased dendritic microtubule associated protein 2 (MAP2) staining in the hippocampus. The diets administered herein were used to examine components from the previous diet and further examine their effects on hippocampal morphology. Specifically, neuronal morphology, dendritic integrity, fatty acid metabolism, microgliosis, and blood vessel structure in the hippocampus and/or adjacent structures were explored. Our results indicate alterations to peripheral and neural systems following each of the diets.
Collapse
Affiliation(s)
- Linnea R Freeman
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | |
Collapse
|
30
|
Maccarrone M, Bernardi G, Agrò AF, Centonze D. Cannabinoid receptor signalling in neurodegenerative diseases: a potential role for membrane fluidity disturbance. Br J Pharmacol 2011; 163:1379-90. [PMID: 21323908 PMCID: PMC3165948 DOI: 10.1111/j.1476-5381.2011.01277.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/13/2011] [Accepted: 01/24/2011] [Indexed: 11/30/2022] Open
Abstract
Type-1 cannabinoid receptor (CB(1)) is the most abundant G-protein-coupled receptor (GPCR) in the brain. CB(1) and its endogenous agonists, the so-called 'endocannabinoids (eCBs)', belong to an ancient neurosignalling system that plays important functions in neurodegenerative and neuroinflammatory disorders like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. For this reason, research on the therapeutic potential of drugs modulating the endogenous tone of eCBs is very intense. Several GPCRs reside within subdomains of the plasma membranes that contain high concentrations of cholesterol: the lipid rafts. Here, the hypothesis that changes in membrane fluidity alter function of the endocannabinoid system, as well as progression of particular neurodegenerative diseases, is described. To this end, the impact of membrane cholesterol on membrane properties and hence on neurodegenerative diseases, as well as on CB(1) signalling in vitro and on CB(1) -dependent neurotransmission within the striatum, is discussed. Overall, present evidence points to the membrane environment as a critical regulator of signal transduction triggered by CB(1) , and calls for further studies aimed at better clarifying the contribution of membrane lipids to eCBs signalling. The results of these investigations might be exploited also for the development of novel therapeutics able to combat disorders associated with abnormal activity of CB(1).
Collapse
Affiliation(s)
- M Maccarrone
- Department of Biomedical Sciences, University of Teramo, Teramo 64100, Italy.
| | | | | | | |
Collapse
|
31
|
Chen X, Wagener JF, Morgan DH, Hui L, Ghribi O, Geiger JD. Endolysosome mechanisms associated with Alzheimer's disease-like pathology in rabbits ingesting cholesterol-enriched diet. J Alzheimers Dis 2011; 22:1289-303. [PMID: 20930277 DOI: 10.3233/jad-2010-101323] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is characterized clinically by progressive disturbances in memory, judgment, reasoning, and olfaction, and pathologically by loss of synaptic integrity, extracellular accumulations of amyloid-β (Aβ) containing plaques, and intraneuronal tangles composed of hyperphosphorylated tau. Endolysosome dysfunction is one of the earliest pathological features of AD and cholesterol, a known risk factor for sporadic AD, is up-taken into neurons via receptor-mediated endocytosis. Accordingly, we determined the extent to which endolysosome dysfunction is associated with pathological features observed in rabbits fed cholesterol-enriched diet; a well-characterized model of sporadic AD. Olfactory bulbs were taken from rabbits fed for 12 weeks a diet enriched with 2% cholesterol and endolysosome morphology and function as well as AD-like pathology were investigated using enzyme activity measurements, immunoblotting and immunostaining techniques. In olfactory bulbs of rabbits fed cholesterol-enriched diet, we observed enlarged endolysosomes containing increased accumulations of ApoB containing cholesterol and increased accumulations of synaptophysin, Aβ, and phosphorylated tau. The cholesterol-enriched diet also significantly decreased specific enzyme activities of the endolysosome enzymes acid phosphatase and cathepsin D. Decreased synaptic area was present in olfactory bulbs of cholesterol-fed rabbits as indicated by significant decreases in protein expression levels of the synaptic area marker protein synaptophysin. Our results suggest strongly that elevated circulating cholesterol plays an important role in the pathogenesis of AD, and that alterations in endolysosome structure and function are associated with cholesterol diet-induced AD-like pathology.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | | | | | | | | | | |
Collapse
|
32
|
Freeman LR, Small BJ, Bickford PC, Umphlet C, Granholm AC. A high-fat/high-cholesterol diet inhibits growth of fetal hippocampal transplants via increased inflammation. Cell Transplant 2011; 20:1499-514. [PMID: 21396159 PMCID: PMC4830280 DOI: 10.3727/096368910x557281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A diet containing high levels of saturated fat and cholesterol is detrimental to many aspects of health and is known to lead to obesity, metabolic syndrome, heart disease, diabetes, and cancer. However, the effects of a diet rich in saturated fat and cholesterol on the brain are not currently well understood. In order to determine direct effects of a high saturated fat and cholesterol diet upon fetal hippocampal tissue, we transplanted hippocampal grafts from embryonic day 18 rats to the anterior eye chamber of 16-month-old host animals that were fed either a normal rat chow diet or a 10% hydrogenated coconut oil + 2% cholesterol diet (HFHC diet) for 8 weeks. One eye per rat received topical application of an IL-1 receptor antagonist (IL-1Ra, Kineret®) and the other served as a saline control. Results revealed that the HFHC diet led to a marked reduction in hippocampal transplant growth, and detrimental effects of the diet were alleviated by the IL-1 receptor antagonist IL-1Ra. Graft morphology demonstrated that the HFHC diet reduced organotypical development of the hippocampal neuronal cell layers, which was also alleviated by IL-1Ra. Finally, grafts were evaluated with markers for glucose transporter expression, astrocytes, and activated microglia. Our results demonstrate significant effects of the HFHC diet on hippocampal morphology, including elevated microglial activation and reduced neuronal development. IL-1Ra largely blocked the detrimental effects of this diet, suggesting a potential use for this agent in neurological disorders involving neuroinflammation.
Collapse
Affiliation(s)
- L R Freeman
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | | | |
Collapse
|
33
|
Neuronal LRP1 knockout in adult mice leads to impaired brain lipid metabolism and progressive, age-dependent synapse loss and neurodegeneration. J Neurosci 2011; 30:17068-78. [PMID: 21159977 DOI: 10.1523/jneurosci.4067-10.2010] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The vast majority of Alzheimer's disease (AD) cases are late onset with progressive synapse loss and neurodegeneration. Although the amyloid hypothesis has generated great insights into the disease mechanism, several lines of evidence indicate that other risk factors might precondition the brain to amyloid toxicity. Here, we show that the deletion of a major lipoprotein receptor, low-density lipoprotein receptor-related protein 1 (LRP1), in forebrain neurons in mice leads to a global defect in brain lipid metabolism characterized by decreased brain levels of cholesterol, sulfatide, galactosylceramide, and triglyceride. These lipid deficits correlate with progressive, age-dependent dendritic spine degeneration, synapse loss, neuroinflammation, memory loss, and eventual neurodegeneration. We further show that the levels of glutamate receptor subunits NMDA receptor 1 and Glu receptor 1 are selectively reduced in LRP1 forebrain knock-out mice and in LRP1 knockdown neurons, which is partially rescued by restoring neuronal cholesterol. Together, these studies support a critical role for LRP1 in maintaining brain lipid homeostasis and associated synaptic and neuronal integrity, and provide important insights into the pathophysiological mechanisms in AD.
Collapse
|
34
|
Chen X, Ghribi O, Geiger JD. Caffeine protects against disruptions of the blood-brain barrier in animal models of Alzheimer's and Parkinson's diseases. J Alzheimers Dis 2010; 20 Suppl 1:S127-41. [PMID: 20164568 DOI: 10.3233/jad-2010-1376] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sporadic Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases and as such they represent major public health problems. Finding effective treatments for AD and PD represents an unmet and elusive goal largely because these diseases are chronic and progressive, and have a complicated and ill-understood pathogenesis. Although the underlying mechanisms are not fully understood, caffeine, the most commonly ingested psychoactive drug in the world, has been shown in human and animal studies to be protective against AD and PD. One mechanism implicated in the pathogenesis of AD and PD is blood-brain barrier (BBB) dysfunction and we reported recently that caffeine exerts protective effects against AD and PD at least in part by keeping the BBB intact. The present review focuses on the role of BBB dysfunction in the pathogenesis of AD and PD, caffeine's protective effects against AD and PD, and potential mechanisms whereby caffeine protects against BBB leakage.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | | | | |
Collapse
|
35
|
Abstract
AD (Alzheimer's disease) is a progressive neurodegenerative disease of unknown origin. Despite questions as to the underlying cause(s) of this disease, shared risk factors for both AD and atherosclerotic cardiovascular disease indicate that vascular mechanisms may critically contribute to the development and progression of both AD and atherosclerosis. An increased risk of developing AD is linked to the presence of the apoE4 (apolipoprotein E4) allele, which is also strongly associated with increased risk of developing atherosclerotic cardiovascular disease. Recent studies also indicate that cardiovascular risk factors, including elevated blood cholesterol and triacylglycerol (triglyceride), increase the likelihood of AD and vascular dementia. Lipids and lipoproteins in the circulation interact intimately with the cerebrovasculature, and may have important effects on its constituent brain microvascular endothelial cells and the adjoining astrocytes, which are components of the neurovascular unit. The present review will examine the potential mechanisms for understanding the contributions of vascular factors, including lipids, lipoproteins and cerebrovascular Abeta (amyloid beta), to AD, and suggest therapeutic strategies for the attenuation of this devastating disease process. Specifically, we will focus on the actions of apoE, TGRLs (triacylglycerol-rich lipoproteins) and TGRL lipolysis products on injury of the neurovascular unit and increases in blood-brain barrier permeability.
Collapse
|
36
|
Gershkovich P, Sivak O, Sharma A, Barr AM, Procyshyn R, Wasan KM. Effect of hypertriglyceridemia on the pharmacokinetics and blood-brain barrier penetration of clozapine and norclozapine following administration to rats. Eur Neuropsychopharmacol 2010; 20:545-52. [PMID: 20163937 DOI: 10.1016/j.euroneuro.2010.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 12/31/2009] [Accepted: 01/21/2010] [Indexed: 10/19/2022]
Abstract
There is a long-term discussion in the literature concerning the possible link between the improved efficacy of clozapine treatment and elevated plasma triglyceride levels, but no mechanistic studies have been performed to date. The aim of this work was to investigate whether the postprandial hypertriglyceridemia affects the pharmacokinetics and brain distribution of clozapine and norclozapine. Experimental hypertriglyceridemia in rats was induced by oral administration of peanut oil and the pharmacokinetic parameters and brain penetration of clozapine and norclozapine following administration of clozapine were compared to normotriglyceridemic control animals. Moderately increased clearance of clozapine was found in hypertriglyceridemic animals compared to control group. No changes were found in penetration of compounds across the blood-brain barrier (BBB). Taken together, the results do not support the hypothesis that hypertriglyceridemia improves the effect of clozapine by altered pharmacokinetics of clozapine and norclozapine and their increased penetration across the BBB.
Collapse
Affiliation(s)
- Pavel Gershkovich
- Division of Pharmaceutics and Biopharmaceutics, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | | | | | |
Collapse
|
37
|
Interplay between cholesterol and drug metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2010; 1814:146-60. [PMID: 20570756 DOI: 10.1016/j.bbapap.2010.05.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/14/2022]
Abstract
Cholesterol biosynthetic and metabolic pathways contain several branching points towards physiologically active molecules, such as coenzyme Q, vitamin D, glucocorticoid and steroid hormones, oxysterols, or bile acids. Sophisticated regulatory mechanisms are involved in maintenance of the homeostasis of not only cholesterol but also other cholesterogenic molecules. In addition to endogenous cues, cholesterol homeostasis needs to accommodate also to exogenous cues that are imported into the body, such as chemicals and medications. Steroid and nuclear receptors together with sterol regulatory element-binding protein (SREBP) mediate the fine tuning of biosynthetic and metabolic routes as well as transports of cholesterol and its derivatives. Similarly, drug/xenobiotic metabolism is the subject to the feedback regulation of cytochrome P450 enzymes and transporters. The regulatory mechanisms that maintain the homeostasis of cholesterogenic molecules and are involved in drug metabolism share similarities. Cholesterol and cholesterogenic compounds (bile acids, glucocorticoids, vitamin D, etc.) regulate the xenosensor signaling in drug-mediated induction of the major drug-metabolizing cytochrome P450 enzymes. The key cellular receptors, pregnane X receptor (PXR), constitutive androstane receptor (CAR), vitamin D receptor (VDR), and glucocorticoid receptor (GR) provide a functional cross-talk between the pathways maintaining cholesterol homeostasis and controlling the expression of drug-metabolizing enzymes. These receptors serve as metabolic sensors, resulting in a coordinate regulation of cholesterogenic compounds metabolism and of the defense against xenobiotic and endobiotic toxicity. Herein we present a comprehensive review of functional interactions between cholesterol homeostasis and drug metabolism involving the main nuclear and steroid receptors.
Collapse
|
38
|
Lee JS, Cho WJ, Shin L, Jena BP. Involvement of cholesterol in synaptic vesicle swelling. Exp Biol Med (Maywood) 2010; 235:470-7. [PMID: 20407079 DOI: 10.1258/ebm.2010.009259] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Studies demonstrate that cholesterol plays a critical role in the regulation of neurotransmitter release and that secretory vesicle swelling is a requirement for the regulated expulsion of intravesicular contents during cell secretion. In view of this, the involvement of cholesterol in synaptic vesicle swelling was hypothesized and tested in the present study, using isolated synaptic vesicles from rat brain and the determination of their swelling competency in the presence and absence of cholesterol. The involvement of the water channel aquaporin-6 (AQP-6) and proton pump vH(+)-ATPase in GTP-G(alpha o)-mediated synaptic vesicle swelling has been reported previously. Mastoparan, the amphiphilic tetradecapeptide from wasp venom, known to activate the GTPase activity of G(alpha o/i) proteins, stimulates synaptic vesicle swelling in the presence of GTP. In the current study, using nanometer-scale precision measurements of isolated synaptic vesicles, we report for the first time that depletion of cholesterol from synaptic vesicle membrane results in a significant loss of GTP-mastoparan-stimulable synaptic vesicle swelling. In contrast, incorporation of cholesterol into the synaptic vesicle membrane potentiates GTP-mastoparan-stimulable vesicle swelling. Our study further demonstrates that this effect of cholesterol is due, in part, to its involvement in the interactions between AQP-6, vH(+)-ATPase and the GTP-binding G(alpha o) protein at the synaptic vesicle membrane.
Collapse
Affiliation(s)
- Jin-Sook Lee
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Bilayer synthesis during membrane biogenesis involves the concerted assembly of multiple lipid species, requiring coordination of the level of lipid synthesis, uptake, turnover, and subcellular distribution. In this review, we discuss some of the salient conclusions regarding the coordination of lipid synthesis that have emerged from work in mammalian and yeast cells. The principal instruments of global control are a small number of transcription factors that target a wide range of genes encoding enzymes that operate in a given metabolic pathway. Critical in mammalian cells are sterol regulatory element binding proteins (SREBPs) that stimulate expression of genes for the uptake and synthesis of cholesterol and fatty acids. From work with Saccharomyces cerevisiae, much has been learned about glycerophospholipid and ergosterol regulation through Ino2p/Ino4p and Upc2p transcription factors, respectively. Lipid supply is fine-tuned through a multitude of negative feedback circuits initiated by both end products and intermediates of lipid synthesis pathways. Moreover, there is evidence that the diversity of membrane lipids is maintained through cross-regulatory effects, whereby classes of lipids activate the activity of enzymes operating in another metabolic branch.
Collapse
Affiliation(s)
- Axel Nohturfft
- Molecular and Metabolic Signalling Centre, Division of Basic Medical Sciences, St. George's University of London, London, SW17 0RE United Kingdom.
| | | |
Collapse
|
40
|
Adibhatla RM, Hatcher JF. Lipid oxidation and peroxidation in CNS health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2010; 12:125-69. [PMID: 19624272 DOI: 10.1089/ars.2009.2668] [Citation(s) in RCA: 317] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) are produced at low levels in mammalian cells by various metabolic processes, such as oxidative phosphorylation by the mitochondrial respiratory chain, NAD(P)H oxidases, and arachidonic acid oxidative metabolism. To maintain physiological redox balance, cells have endogenous antioxidant defenses regulated at the transcriptional level by Nrf2/ARE. Oxidative stress results when ROS production exceeds the cell's ability to detoxify ROS. Overproduction of ROS damages cellular components, including lipids, leading to decline in physiological function and cell death. Reaction of ROS with lipids produces oxidized phospholipids, which give rise to 4-hydroxynonenal, 4-oxo-2-nonenal, and acrolein. The brain is susceptible to oxidative damage due to its high lipid content and oxygen consumption. Neurodegenerative diseases (AD, ALS, bipolar disorder, epilepsy, Friedreich's ataxia, HD, MS, NBIA, NPC, PD, peroxisomal disorders, schizophrenia, Wallerian degeneration, Zellweger syndrome) and CNS traumas (stroke, TBI, SCI) are problems of vast clinical importance. Free iron can react with H(2)O(2) via the Fenton reaction, a primary cause of lipid peroxidation, and may be of particular importance for these CNS injuries and disorders. Cholesterol is an important regulator of lipid organization and the precursor for neurosteroid biosynthesis. Atherosclerosis, the major risk factor for ischemic stroke, involves accumulation of oxidized LDL in the arteries, leading to foam cell formation and plaque development. This review will discuss the role of lipid oxidation/peroxidation in various CNS injuries/disorders.
Collapse
Affiliation(s)
- Rao Muralikrishna Adibhatla
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792-3232, USA.
| | | |
Collapse
|
41
|
Liu JP, Tang Y, Zhou S, Toh BH, McLean C, Li H. Cholesterol involvement in the pathogenesis of neurodegenerative diseases. Mol Cell Neurosci 2010; 43:33-42. [DOI: 10.1016/j.mcn.2009.07.013] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 07/29/2009] [Accepted: 07/29/2009] [Indexed: 11/27/2022] Open
|
42
|
Vik-Mo AO, Fernø J, Skrede S, Steen VM. Psychotropic drugs up-regulate the expression of cholesterol transport proteins including ApoE in cultured human CNS- and liver cells. BMC Pharmacol 2009; 9:10. [PMID: 19715613 PMCID: PMC2753324 DOI: 10.1186/1471-2210-9-10] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 08/29/2009] [Indexed: 01/20/2023] Open
Abstract
Background Disturbances in lipid homeostasis and myelination have been proposed in the pathophysiology of schizophrenia and bipolar disorder. We have previously shown that several antipsychotic and antidepressant drugs increase lipid biosynthesis through activation of the Sterol Regulatory Element-Binding Protein (SREBP) transcription factors, which control the expression of numerous genes involved in fatty acid and cholesterol biosynthesis. The aim of the present proof-of-principle study was to investigate whether such drugs also affect lipid transport and export pathways in cultured human CNS and liver cells. Results Quantitative PCR and immunoblotting were used to determine the level of lipid transport genes in human glioblastoma (GaMg) exposed to clozapine, olanzapine, haloperidol or imipramine. The effect of some of these drugs was also investigated in human astrocytoma (CCF-STTG1), neuroblastoma (SH-SY5Y) and hepatocellular carcinoma (HepG2) cells. We found significant transcriptional changes of cholesterol transport genes (ApoE, ABCA1, NPC1, NPC2, NPC1L1), which are predominantly controlled by the Liver X receptor (LXR) transcription factor. The up-regulation was observed after 24 to 48 hours of drug exposure, which is markedly delayed as compared to the drug-induced SREBP-controlled stimulation of lipid biosynthesis seen after 6 hours. Conclusion Our data show that stimulation of cellular lipid biosynthesis by amphiphilic psychotropic drugs is followed by a transcriptional activation of cholesterol transport and efflux pathways. Such effects may be relevant for both therapeutic effects and metabolic adverse effects of psychotropic drugs.
Collapse
Affiliation(s)
- Audun O Vik-Mo
- Department of Clinical Medicine, University of Bergen, Dr Einar Martens' Research Group for Biological Psychiatry and Bergen Mental Health Research Center, Norway.
| | | | | | | |
Collapse
|
43
|
Kaur D, Rajagopalan S, Andersen JK. Chronic expression of H-ferritin in dopaminergic midbrain neurons results in an age-related expansion of the labile iron pool and subsequent neurodegeneration: implications for Parkinson's disease. Brain Res 2009; 1297:17-22. [PMID: 19699718 DOI: 10.1016/j.brainres.2009.08.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 08/08/2009] [Accepted: 08/13/2009] [Indexed: 10/20/2022]
Abstract
While ferritin elevation within dopaminergic (DA) neurons of the substantia nigra (SN) is protective against neurodegeneration elicited by two toxin models of Parkinson's disease (PD), MPTP and paraquat, in young animals, its prolonged elevation results in a selective age-related neurodegeneration. A similar age-related neurodegeneration has been reported in iron regulatory protein 2-deficient (IRP2 -/-) mice coinciding with increased ferritin levels within degenerating neurons. This has been speculated to be due to subsequent reductions in the labile iron pool (LIP) needed for the synthesis of iron-sulfur-containing enzymes. In order to assess whether LIP reduction is responsible for age-related neurodegeneration in our ferritin transgenics, we examined LIP levels in ferritin-expressing transgenics with increasing age. While LIP levels were reduced within DA SN nerve terminals isolated from young ferritin transgenics compared to wildtype littermate controls, they were found to be increased in older transgenic animals at the age at which selective neurodegeneration is first noted. Furthermore, administration of the bioavailable iron chelator, clioquinol (CQ), to older mice was found to protect against both increased LIP and subsequent dopaminergic neurodegeneration. This suggests that age-related neurodegeneration in these mice is likely due to increased iron availability rather than its reduction. This may have important implications for PD and other related neurodegenerative conditions in which iron and ferritin have been implicated.
Collapse
Affiliation(s)
- Deepinder Kaur
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, CA 94945, USA
| | | | | |
Collapse
|
44
|
Mental retardation linked to mutations in the HSD17B10 gene interfering with neurosteroid and isoleucine metabolism. Proc Natl Acad Sci U S A 2009; 106:14820-4. [PMID: 19706438 DOI: 10.1073/pnas.0902377106] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in the HSD17B10 gene were identified in two previously described mentally retarded males. A point mutation c.776G>C was found from a survivor (SV), whereas a potent mutation, c.419C>T, was identified in another deceased case (SF) with undetectable hydroxysteroid (17beta) dehydrogenase 10 (HSD10) activity. Protein levels of mutant HSD10(R130C) in patient SF and HSD10(E249Q) in patient SV were about half that of HSD10 in normal controls. The E249Q mutation appears to affect HSD10 subunit interactions, resulting in an allosteric regulatory enzyme. For catalyzing the oxidation of allopregnanolone by NAD+ the Hill coefficient of the mutant enzyme is approximately 1.3. HSD10(E249Q) was unable to catalyze the dehydrogenation of 2-methyl-3-hydroxybutyryl-CoA and the oxidation of allopregnanolone, a positive modulator of the gamma-aminobutyric acid type A receptor, at low substrate concentrations. Neurosteroid homeostasis is critical for normal cognitive development, and there is increasing evidence that a blockade of isoleucine catabolism alone does not commonly cause developmental disabilities. The results support the theory that an imbalance in neurosteroid metabolism could be a major cause of the neurological handicap associated with hydroxysteroid (17beta) dehydrogenase 10 deficiency.
Collapse
|
45
|
Walkley SU, Vanier MT. Secondary lipid accumulation in lysosomal disease. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1793:726-36. [PMID: 19111580 PMCID: PMC4382014 DOI: 10.1016/j.bbamcr.2008.11.014] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 11/11/2008] [Accepted: 11/28/2008] [Indexed: 01/22/2023]
Abstract
Lysosomal diseases are inherited metabolic disorders caused by defects in a wide spectrum of lysosomal and a few non-lysosomal proteins. In most cases a single type of primary storage material is identified, which has been used to name and classify the disorders: hence the terms sphingolipidoses, gangliosidoses, mucopolysaccharidoses, glycoproteinoses, and so forth. In addition to this primary storage, however, a host of secondary storage products can also be identified, more often than not having no direct link to the primary protein defect. Lipids - glycosphingolipids and phospholipids, as well as cholesterol - are the most ubiquitous and best studied of these secondary storage materials. While in the past typically considered nonspecific and nonconsequential features of these diseases, newer studies suggest direct links between secondary storage and disease pathogenesis and support the view that understanding all aspects of this sequestration process will provide important insights into the cell biology and treatment of lysosomal disease.
Collapse
Affiliation(s)
- Steven U Walkley
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA.
| | | |
Collapse
|
46
|
Expression of cholesterol homeostasis genes in the brain of the male rat is affected by age and dietary restriction. Biogerontology 2009; 10:735-45. [DOI: 10.1007/s10522-009-9220-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/26/2009] [Indexed: 10/21/2022]
|
47
|
Abstract
PURPOSE OF REVIEW Aberrations in cerebral cholesterol homeostasis can lead to severe neurological diseases and have been linked to Alzheimer's disease. Many proteins involved in peripheral cholesterol metabolism are also present in the brain. Yet, brain cholesterol metabolism is very different from that in the remainder of the body. This review reports on present insights into the regulation of cerebral cholesterol homeostasis, focusing on cholesterol trafficking between astrocytes and neurons. RECENT FINDINGS Astrocytes are a major site of cholesterol synthesis. They secrete cholesterol in the form of apolipoprotein E-containing HDL-like particles. After birth, neurons are thought to reduce their cholesterol synthesis and rely predominantly on astrocytes for their cholesterol supply. How exactly neurons regulate their cholesterol supply is largely unknown. A role for the brain-specific cholesterol metabolite, 24(S)-hydroxycholesterol, in this process was recently proposed. Recent findings strengthen the link between brain cholesterol metabolism and factors involved in synaptic plasticity, a process essential for learning and memory functions, as well as regeneration, which are affected in Alzheimer's disease. SUMMARY Insight into the regulation of cerebral cholesterol homeostasis will provide possibilities to modulate the key steps involved and may lead to the development of therapies for the prevention as well as treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Monique Mulder
- Department of Internal Medicine and Division of Pharmacology, Vascular and Metabolic diseases, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
48
|
Modifications of the expression of genes involved in cerebral cholesterol metabolism in the rat following chronic ingestion of depleted uranium. J Mol Neurosci 2008; 38:159-65. [PMID: 18792811 DOI: 10.1007/s12031-008-9145-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 08/25/2008] [Indexed: 10/21/2022]
Abstract
Depleted uranium results from the enrichment of natural uranium for energetic purpose. Its potential dispersion in the environment would set human populations at risk of being contaminated through ingestion. Uranium can build up in the brain and induce behavior disorders. As a major constituent of the myelin sheath, cholesterol is essential to brain function, and several neurological pathologies result from a disruption of cholesterol metabolism. To assess the effect of a chronic contamination with depleted uranium on cerebral cholesterol metabolism, rats were exposed to depleted uranium for 9 months through drinking water at 40 mg/l. The study focuses on gene expression. Cholesterol-catabolizing enzyme CYP46A1 displayed a 39% increase of its messenger RNA (mRNA) level. 3-Hydroxy-3-methylglutamyl CoA synthase gene expression rose from 91%. Concerning cholesterol transport, mRNA levels of scavenger receptor-B1 and adenosine triphosphate-binding cassette transporter A1 increased by 34% and that of apolipoprotein E by 75%. Concerning regulation, gene expression of nuclear receptors peroxisome proliferator-activated receptors alpha and gamma increased by 46% and 36% respectively, whereas that of retinoid-X-receptor decreased by 29%. In conclusion, a chronic internal contamination with depleted uranium does not affect the health status of rats but induces molecular changes in the dynamic equilibrium of the cerebral cholesterol pool.
Collapse
|
49
|
Neuronal loss of Drosophila NPC1a causes cholesterol aggregation and age-progressive neurodegeneration. J Neurosci 2008; 28:6569-82. [PMID: 18579730 DOI: 10.1523/jneurosci.5529-07.2008] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The mistrafficking and consequent cytoplasmic accumulation of cholesterol and sphingolipids is linked to multiple neurodegenerative diseases. One class of disease, the sphingolipid storage diseases, includes Niemann-Pick disease type C (NPC), caused predominantly (95%) by mutation of the NPC1 gene. A disease model has been established through mutation of Drosophila NPC1a (dnpc1a). Null mutants display early lethality attributable to loss of cholesterol-dependent ecdysone steroid hormone production. Null mutants rescued to adults by restoring ecdysone production mimic human NPC patients with progressive motor defects and reduced life spans. Analysis of dnpc1a null brains shows elevated overall cholesterol levels and progressive accumulation of filipin-positive cholesterol aggregates within brain and retina, as well as isolated cultured brain neurons. Ultrastructural imaging of dnpc1a mutant brains reveals age-progressive accumulation of striking multilamellar and multivesicular organelles, preceding the onset of neurodegeneration. Consistently, electroretinogram recordings show age-progressive loss of phototransduction and photoreceptor synaptic transmission. Early lethality, movement impairments, neuronal cholesterol deposits, accumulation of multilamellar bodies, and age-dependent neurodegeneration are all rescued by targeted neuronal expression of a wild-type dnpc1a transgene. Interestingly, targeted expression of dnpc1a in glia also provides limited rescue of adult lethality. Generation of dnpc1a null mutant neuron clones in the brain reveals cell-autonomous requirements for dNPC1a in cholesterol and membrane trafficking. These data demonstrate a requirement for dNPC1a in the maintenance of neuronal function and viability and show that loss of dNPC1a in neurons mimics the human neurodegenerative condition.
Collapse
|
50
|
The Role and Metabolism of Sulfatide in the Nervous System. Mol Neurobiol 2008; 37:93-103. [DOI: 10.1007/s12035-008-8022-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/09/2008] [Indexed: 12/16/2022]
|