1
|
Devinat M, Thevenard-Devy J, Ghilane F, Devy J, Chazee L, Terryn C, Duca L, Devarenne-Charpentier E, El Btaouri H. Xanthohumol Sensitizes Melanoma Cells to Vemurafenib by Lowering Membrane Cholesterol and Increasing Membrane Fluidity. Int J Mol Sci 2025; 26:2290. [PMID: 40076912 PMCID: PMC11901044 DOI: 10.3390/ijms26052290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Chemoresistance remains one of the major obstacles to cancer treatment. The search for specific molecules that could improve cancer treatment has become one of the objectives of biomedical research. Identifying new natural molecules to enhance chemotherapy treatment or improve sensitization to conventional therapies has become a key objective. Here, we evaluated the effect of Xanthohumol (XN) extracted from hop on SKMEL-28 melanoma cells and their sensitization to vemurafenib (VEM) treatment. We measured the XN effect on cell viability and apoptosis. We also assessed the effect of XN on membrane fluidity and membrane cholesterol levels. Finally, we studied the impact of XN on cell sensitization to VEM. Here, we showed that XN reduced SKMEL-28 cell viability through an apoptotic mechanism. Our results demonstrated the potential role of XN in sensitizing cancer cells to VEM with a less toxic effect on non-tumor cells. A study of XN's molecular mechanism showed that XN was able to induce cholesterol depletion and increased fluidity in SKMEL-28 cancer cells. This leads to an increase in VEM incorporation. Here, we describe the importance of the strategy to modulate membrane fluidity by XN in order to significantly improve anticancer therapy.
Collapse
Affiliation(s)
- Marine Devinat
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Jessica Thevenard-Devy
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Fatiha Ghilane
- Laboratoire de Biologie des Pathologies Humaines, Université Mohammed V de Rabat, 4 Avenue Ibn Battouta, Rabat B.P. 1014 RP, Morocco;
| | - Jerome Devy
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Lise Chazee
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Christine Terryn
- Plateau Technique en Imagerie Cellulaire et Tissulaire (PICT) Pôle Santé, UFR Pharmacie, Université de Reims Champagne Ardenne, 51 Rue Cognacq Jay, 51096 Reims, France;
| | - Laurent Duca
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Emmanuelle Devarenne-Charpentier
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Hassan El Btaouri
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| |
Collapse
|
2
|
Valencia-Olvera AC, Balu D, Faulk N, Amiridis A, Wang Y, Pham C, Avila-Munoz E, York JM, Thatcher GRJ, LaDu MJ. Inhibition of ACAT as a Therapeutic Target for Alzheimer's Disease Is Independent of ApoE4 Lipidation. Neurotherapeutics 2023; 20:1120-1137. [PMID: 37157042 PMCID: PMC10457278 DOI: 10.1007/s13311-023-01375-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 05/10/2023] Open
Abstract
APOE4, encoding apolipoprotein E4 (apoE4), is the greatest genetic risk factor for Alzheimer's disease (AD), compared to the common APOE3. While the mechanism(s) underlying APOE4-induced AD risk remains unclear, increasing the lipidation of apoE4 is an important therapeutic target as apoE4-lipoproteins are poorly lipidated compared to apoE3-lipoproteins. ACAT (acyl-CoA: cholesterol-acyltransferase) catalyzes the formation of intracellular cholesteryl-ester droplets, reducing the intracellular free cholesterol (FC) pool. Thus, inhibiting ACAT increases the FC pool and facilitates lipid secretion to extracellular apoE-containing lipoproteins. Previous studies using commercial ACAT inhibitors, including avasimibe (AVAS), as well as ACAT-knock out (KO) mice, exhibit reduced AD-like pathology and amyloid precursor protein (APP) processing in familial AD (FAD)-transgenic (Tg) mice. However, the effects of AVAS with human apoE4 remain unknown. In vitro, AVAS induced apoE efflux at concentrations of AVAS measured in the brains of treated mice. AVAS treatment of male E4FAD-Tg mice (5xFAD+/-APOE4+/+) at 6-8 months had no effect on plasma cholesterol levels or distribution, the original mechanism for AVAS treatment of CVD. In the CNS, AVAS reduced intracellular lipid droplets, indirectly demonstrating target engagement. Surrogate efficacy was demonstrated by an increase in Morris water maze measures of memory and postsynaptic protein levels. Amyloid-beta peptide (Aβ) solubility/deposition and neuroinflammation were reduced, critical components of APOE4-modulated pathology. However, there was no increase in apoE4 levels or apoE4 lipidation, while amyloidogenic and non-amyloidogenic processing of APP were significantly reduced. This suggests that the AVAS-induced reduction in Aβ via reduced APP processing was sufficient to reduce AD pathology, as apoE4-lipoproteins remained poorly lipidated.
Collapse
Affiliation(s)
- Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Naomi Faulk
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | | | - Yueting Wang
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612 USA
- Present Address: AbbVie Inc., 1 N. Waukegan Road, North Chicago, IL 60064 USA
| | - Christine Pham
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Eva Avila-Munoz
- Syneos Health, Av. Gustavo Baz 309, La Loma, Tlalnepantla de Baz, 54060 Mexico
| | - Jason M. York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Gregory R. J. Thatcher
- Department of Pharmacology & Toxicology, University of Arizona, 1703 E Mabel St., Tucson, AZ 85721 USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
3
|
Sahin I, Ceylan Ç, Bayraktar O. Ruscogenin interacts with DPPC and DPPG model membranes and increases the membrane fluidity: FTIR and DSC studies. Arch Biochem Biophys 2023; 733:109481. [PMID: 36522815 DOI: 10.1016/j.abb.2022.109481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022]
Abstract
Ruscogenin, a kind of steroid saponin, has been shown to have significant anti-oxidant, anti-inflammatory, and anti-thrombotic characteristics. Furthermore, it has the potential to be employed as a medicinal medication to treat a variety of acute and chronic disorders. The interaction of a drug molecule with cell membranes can help to elucidate its system-wide protective and therapeutic effects, and it's also important for its pharmacological activity. The molecular mechanism by which ruscogenin affects membrane architecture is still a mystery. Ruscogenin's interaction with zwitterionic dipalmitoyl phosphatidylcholine (DPPC) and anionic dipalmitoyl phosphatidylglycerol (DPPG) multilamellar vesicles (MLVs) was studied utilizing two non-invasive approaches, including: Fourier Transform Infrared (FTIR) spectroscopy and Differential Scanning Calorimetry. Ruscogenin caused considerable alterations in the phase transition profile, order, dynamics and hydration state of head groups and glycerol backbone of DPPC and DPPG MLVs at all concentrations. The DSC results indicated that the presence of ruscogenin decreased the main phase transition temperature (Tm) and enthalpy (ΔH) values of both membranes and increased half height width of the main transition (ΔT1/2). The FTIR results demonstrated that all concentrations (1, 3, 6, 9, 15, 24 and 30 mol percent) of ruscogenin disordered the DPPC MLVs both in the gel and liquid crystalline phases while it increased the order of DPPG MLVs in the liquid crystalline phase. Moreover, ruscogenin caused an increase in the dynamics of DPPC and DPPG MLVs in both phases. Additionally, it enhanced the hydration of the head groups of lipids and the surrounding water molecules implying ruscogenin to interact strongly with both zwitterionic and charged model membranes.
Collapse
Affiliation(s)
- Ipek Sahin
- Department of Physics, Faculty of Science, Ege University, 35100, Bornova, İzmir, Turkey.
| | - Çağatay Ceylan
- Department of Food Engineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, Turkey
| | - Oguz Bayraktar
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100, Bornova, İzmir, Turkey
| |
Collapse
|
4
|
Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer's Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor α. Cells 2020; 9:E1215. [PMID: 32422896 PMCID: PMC7290654 DOI: 10.3390/cells9051215] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Mutations in genes encoding proteins involved in amyloid-β peptide (Aβ) production are responsible for inherited AD cases. The amyloid cascade hypothesis was proposed to explain the pathogeny. Despite the fact that Aβ is considered as the main culprit of the pathology, most clinical trials focusing on Aβ failed and suggested that earlier interventions are needed to influence the course of AD. Therefore, identifying risk factors that predispose to AD is crucial. Among them, the epsilon 4 allele of the apolipoprotein E gene that encodes the major brain lipid carrier and metabolic disorders such as obesity and type 2 diabetes were identified as AD risk factors, suggesting that abnormal lipid metabolism could influence the progression of the disease. Among lipids, fatty acids (FAs) play a fundamental role in proper brain function, including memory. Peroxisome proliferator-activated receptor α (PPARα) is a master metabolic regulator that regulates the catabolism of FA. Several studies report an essential role of PPARα in neuronal function governing synaptic plasticity and cognition. In this review, we explore the implication of lipid metabolism in AD, with a special focus on PPARα and its potential role in AD therapy.
Collapse
Affiliation(s)
- Francisco Sáez-Orellana
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Jean-Noël Octave
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Nathalie Pierrot
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| |
Collapse
|
5
|
Rad SK, Arya A, Karimian H, Madhavan P, Rizwan F, Koshy S, Prabhu G. Mechanism involved in insulin resistance via accumulation of β-amyloid and neurofibrillary tangles: link between type 2 diabetes and Alzheimer's disease. Drug Des Devel Ther 2018; 12:3999-4021. [PMID: 30538427 PMCID: PMC6255119 DOI: 10.2147/dddt.s173970] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The pathophysiological link between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) has been suggested in several reports. Few findings suggest that T2DM has strong link in the development process of AD, and the complete mechanism is yet to be revealed. Formation of amyloid plaques (APs) and neurofibrillary tangles (NFTs) are two central hallmarks in the AD. APs are the dense composites of β-amyloid protein (Aβ) which accumulates around the nerve cells. Moreover, NFTs are the twisted fibers containing hyperphosphorylated tau proteins present in certain residues of Aβ that build up inside the brain cells. Certain factors contribute to the aetiogenesis of AD by regulating insulin signaling pathway in the brain and accelerating the formation of neurotoxic Aβ and NFTs via various mechanisms, including GSK3β, JNK, CamKII, CDK5, CK1, MARK4, PLK2, Syk, DYRK1A, PPP, and P70S6K. Progression to AD could be influenced by insulin signaling pathway that is affected due to T2DM. Interestingly, NFTs and APs lead to the impairment of several crucial cascades, such as synaptogenesis, neurotrophy, and apoptosis, which are regulated by insulin, cholesterol, and glucose metabolism. The investigation of the molecular cascades through insulin functions in brain contributes to probe and perceive progressions of diabetes to AD. This review elaborates the molecular insights that would help to further understand the potential mechanisms linking T2DM and AD.
Collapse
Affiliation(s)
- Sima Kianpour Rad
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia,
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), Bukit Gambir, Gelugor, Pulau Pinang, Malaysia,
| | - Hamed Karimian
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia,
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Farzana Rizwan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Shajan Koshy
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Girish Prabhu
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
6
|
Liehn EA, Ponomariov V, Diaconu R, Streata I, Ioana M, Crespo-Avilan GE, Hernández-Reséndiz S, Cabrera-Fuentes HA. Apolipoprotein E in Cardiovascular Diseases: Novel Aspects of an Old-fashioned Enigma. Arch Med Res 2018; 49:522-529. [PMID: 30213474 DOI: 10.1016/j.arcmed.2018.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022]
Abstract
The presence of different APOE isoforms represents a well-known risk factor for cardiovascular diseases. Besides the pleiotropic effects of APOE polymorphism on heart and neurological diseases, this review summarizes the less-known functions of APOE and the possible implications for cardiovascular disorders. Beyond the role as lipid transporting protein, its involvement in lipid membrane homeostasis and signaling, as well as its nuclear transcriptional effects suggests a more complex role of APOE, receiving great interest from researchers and physicians from all medical fields. Due to the presence of different APOE isoforms in human population, understanding APOE's role in pathological processes represents not only a challenge, but a demand for further development of therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Elisa A Liehn
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University, Aachen, Germany; Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania; Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital, Rheinisch Westfälische Technische Hochschule, Aachen, Germany
| | - Victor Ponomariov
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University, Aachen, Germany; Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Rodica Diaconu
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ioana Streata
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Mihai Ioana
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Kazan Federal University, Department of Microbiology, Kazan, Russian Federation; Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Tecnologico de Monterrey, Nuevo Leon, México; Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
7
|
Hunter S, Smailagic N, Brayne C. Dementia Research: Populations, Progress, Problems, and Predictions. J Alzheimers Dis 2018; 64:S119-S143. [DOI: 10.3233/jad-179927] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sally Hunter
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Nadja Smailagic
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
D’Auria L, Reiter C, Ward E, Moyano AL, Marshall MS, Nguyen D, Scesa G, Hauck Z, van Breemen R, Givogri MI, Bongarzone ER. Psychosine enhances the shedding of membrane microvesicles: Implications in demyelination in Krabbe's disease. PLoS One 2017; 12:e0178103. [PMID: 28531236 PMCID: PMC5439731 DOI: 10.1371/journal.pone.0178103] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/06/2017] [Indexed: 12/22/2022] Open
Abstract
In prior studies, our laboratory showed that psychosine accumulates and disrupts lipid rafts in brain membranes of Krabbe’s disease. A model of lipid raft disruption helped explaining psychosine’s effects on several signaling pathways important for oligodendrocyte survival and differentiation but provided more limited insight in how this sphingolipid caused demyelination. Here, we have studied how this cationic inverted coned lipid affects the fluidity, stability and structure of myelin and plasma membranes. Using a combination of cutting-edge imaging techniques in non-myelinating (red blood cell), and myelinating (oligodendrocyte) cell models, we show that psychosine is sufficient to disrupt sphingomyelin-enriched domains, increases the rigidity of localized areas in the plasma membrane, and promotes the shedding of membranous microvesicles. The same physicochemical and structural changes were measured in myelin membranes purified from the mutant mouse Twitcher, a model for Krabbe’s disease. Areas of higher rigidity were measured in Twitcher myelin and correlated with higher levels of psychosine and of myelin microvesiculation. These results expand our previous analyses and support, for the first time a pathogenic mechanism where psychosine’s toxicity in Krabbe disease involves deregulation of cell signaling not only by disruption of membrane rafts, but also by direct local destabilization and fragmentation of the membrane through microvesiculation. This model of membrane disruption may be fundamental to introduce focal weak points in the myelin sheath, and consequent diffuse demyelination in this leukodystrophy, with possible commonality to other demyelinating disorders.
Collapse
Affiliation(s)
- Ludovic D’Auria
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Cory Reiter
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Emma Ward
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Ana Lis Moyano
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Michael S. Marshall
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Duc Nguyen
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Giuseppe Scesa
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Zane Hauck
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago, Illinois, United States of America
| | - Richard van Breemen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago, Illinois, United States of America
| | - Maria I. Givogri
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois, Chicago, Illinois, United States of America
- Departamento de Química Biologica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
9
|
Domanski D, Zegrocka-Stendel O, Perzanowska A, Dutkiewicz M, Kowalewska M, Grabowska I, Maciejko D, Fogtman A, Dadlez M, Koziak K. Molecular Mechanism for Cellular Response to β-Escin and Its Therapeutic Implications. PLoS One 2016; 11:e0164365. [PMID: 27727329 PMCID: PMC5058498 DOI: 10.1371/journal.pone.0164365] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/23/2016] [Indexed: 11/18/2022] Open
Abstract
β-escin is a mixture of triterpene saponins isolated from the horse chestnut seeds (Aesculus hippocastanum L.). The anti-edematous, anti-inflammatory and venotonic properties of β-escin have been the most extensively clinically investigated effects of this plant-based drug and randomized controlled trials have proved the efficacy of β-escin for the treatment of chronic venous insufficiency. However, despite the clinical recognition of the drug its pharmacological mechanism of action still remains largely elusive. To determine the cellular and molecular basis for the therapeutic effectiveness of β-escin we performed discovery and targeted proteomic analyses and in vitro evaluation of cellular and molecular responses in human endothelial cells under inflammatory conditions. Our results demonstrate that in endothelial cells β-escin potently induces cholesterol synthesis which is rapidly followed with marked fall in actin cytoskeleton integrity. The concomitant changes in cell functioning result in a significantly diminished responses to TNF-α stimulation. These include reduced migration, alleviated endothelial monolayer permeability, and inhibition of NFκB signal transduction leading to down-expression of TNF-α-induced effector proteins. Moreover, the study provides evidence for novel therapeutic potential of β-escin beyond the current vascular indications.
Collapse
Affiliation(s)
- Dominik Domanski
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Oliwia Zegrocka-Stendel
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Anna Perzanowska
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Dutkiewicz
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Kowalewska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Miecznikowa 1, 02–096 Warsaw, Poland
| | - Dorota Maciejko
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Anna Fogtman
- Laboratory for Microarray Analysis CORELAB, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Dadlez
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Koziak
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Brambillaa A, Lonati E, Milani C, Rizzo AM, Farina F, Botto L, Masserini M, Palestini P, Bulbarelli A. Ischemic conditions and ß-secretase activation: The impact of membrane cholesterol enrichment as triggering factor in rat brain endothelial cells. Int J Biochem Cell Biol 2016; 69:95-104. [PMID: 27022655 DOI: 10.1016/j.biocel.2015.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Among harmful conditions damaging the blood–brain barrier, cerebral stroke and reperfusion injuries were proposed as contributing factors to Alzheimer's disease etiology. Indeed it was reported that ischemic conditions promote β-amyloid peptide production in brain endothelial cells, although implicated mechanisms are yet not fully understood.Oxidative injury related to ischemia affects membrane-lipids profile by altering their biochemical properties and structural dynamics, which are also believed to play significant role in the amyloid precursor protein processing, suggesting a link between alterations in lipid membrane composition and β-amyloid peptide production enhancement.Using brain microvascular endothelial cells, here we demonstrate how oxygen and glucose deprivation followed by normal conditions restoration, mimicking ischemic environment, increases cell cholesterol amount (+20%), reduces membrane fluidity and results in strong activation (+40%) of β-secretase 1 enzymatic activity. Moreover, we observed an increase of amyloid precursor protein and β-secretase 1 protein levels with altered localization in non-discrete (Triton X-100 soluble) membrane domains, leading to an enhanced production of amyloid precursor protein β-carboxyl-terminal fragment. Therefore, lipid alterations induced by oxygen and glucose deprivation enhance β-secretase 1 activity, favor its proximity to amyloid precursor protein and may concur to increased amyloidogenic cleavage. The latter, represents a detrimental event that may contribute to β-amyloid homeostasis impairment in the brain and to Alzheimer's disease-related BBB dysfunctions.
Collapse
|
11
|
Biswal S, Sharma D, Kumar K, Nag TC, Barhwal K, Hota SK, Kumar B. Global hypoxia induced impairment in learning and spatial memory is associated with precocious hippocampal aging. Neurobiol Learn Mem 2016; 133:157-170. [PMID: 27246251 DOI: 10.1016/j.nlm.2016.05.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/09/2016] [Accepted: 05/27/2016] [Indexed: 01/10/2023]
Abstract
Both chronological aging and chronic hypoxia stress have been reported to cause degeneration of hippocampal CA3 neurons and spatial memory impairment through independent pathways. However, the possible occurrence of precocious biological aging on exposure to single episode of global hypoxia resulting in impairment of learning and memory remains to be established. The present study thus aimed at bridging this gap in existing literature on hypoxia induced biological aging. Male Sprague Dawley rats were exposed to simulated hypobaric hypoxia (25,000ft) for different durations and were compared with aged rats. Behavioral studies in Morris Water Maze showed decline in learning abilities of both chronologically aged as well as hypoxic rats as evident from increased latency and pathlength to reach target platform. These behavioral changes in rats exposed to global hypoxia were associated with deposition of lipofuscin and ultrastructural changes in the mitochondria of hippocampal neurons that serve as hallmarks of aging. A single episode of chronic hypobaric hypoxia exposure also resulted in the up-regulation of pro-aging protein, S100A9 and down regulation of Tau, SNAP25, APOE and Sod2 in the hippocampus similar to that in aged rats indicating hypoxia induced accelerated aging. The present study therefore provides evidence for role of biological aging of hippocampal neurons in hypoxia induced impairment of learning and memory.
Collapse
Affiliation(s)
- Suryanarayan Biswal
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India
| | - Deepti Sharma
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India
| | - Kushal Kumar
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Science, New Delhi, India
| | - Kalpana Barhwal
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India
| | - Sunil Kumar Hota
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India.
| | - Bhuvnesh Kumar
- Defence Institute of High Altitude Research, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir 901205, India
| |
Collapse
|
12
|
Yoon MS, Won KJ, Kim DY, Hwang DI, Yoon SW, Jung SH, Lee KP, Jung D, Choi WS, Kim B, Lee HM. Diminished Lipid Raft SNAP23 Increases Blood Pressure by Inhibiting the Membrane Fluidity of Vascular Smooth-Muscle Cells. J Vasc Res 2016; 52:321-33. [PMID: 26930561 DOI: 10.1159/000443888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/10/2016] [Indexed: 11/19/2022] Open
Abstract
Synaptosomal-associated protein 23 (SNAP23) is involved in microvesicle trafficking and exocytosis in various cell types, but its functional role in blood pressure (BP) regulation has not yet been defined. Here, we found that lipid raft SNAP23 expression was much lower in vascular smooth-muscle cells (VSMCs) from spontaneously hypertensive rats (SHR) than in those from normotensive Wistar-Kyoto (WKY) rats. This led us to investigate the hypothesis that this lower expression may be linked to the spontaneous hypertension found in SHR. The expression level of lipid raft SNAP23 and the fluidity in the plasma membrane of VSMCs were lower in SHR than in WKY rats. Cholesterol content in the VSMC membrane was higher, but the secreted cholesterols found in VSMC-conditioned medium and in the blood serum were lower in SHR than in WKY rats. SNAP23 knockdown in WKY rat VSMCs reduced the membrane fluidity and increased the membrane cholesterol level. Systemic overexpression of SNAP23 in SHR resulted in an increase of cholesterol content in their serum, a decrease in cholesterol in their aorta and the reduction of their BP. Our findings suggest that the low expression of the lipid raft SNAP23 in VSMCs might be a potential cause for the characteristic hypertension of SHR.
Collapse
Affiliation(s)
- Mi So Yoon
- Department of Cosmetic Science, College of Life and Health Sciences, Hoseo University, Asan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Agarwal R, Talwar P, Kushwaha SS, Tripathi CB, Kukreti R. Effect of apolipoprotein E (APO E) polymorphism on leptin in Alzheimer's disease. Ann Indian Acad Neurol 2015; 18:320-6. [PMID: 26425011 PMCID: PMC4564468 DOI: 10.4103/0972-2327.157255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Leptin, a 16 kDa peptide hormone synthesized and secreted specifically from white adipose cells protects neurons against amyloid β-induced toxicity, by increasing Apolipoprotein E (APO E)-dependent uptake of β amyloid into the cells, thereby, protect individuals from developing Alzheimer's disease (AD). The APO E ε4 allele is a known genetic risk factor for AD by accelerating onset. It is estimated that the lifetime risk of developing AD increases to 29% for carriers with one ε4 allele and 9% for those with no ε4 allele. Objectives: To determine the levels of serum leptin, cholesterol, low density lipoprotein (LDL-C), and high density lipoprotein (HDL-C) in the diagnosed cases of AD and the association of them with cognitive decline and Apolipoprotein E (APO E) genotypes in AD. Materials and Methods: Serum levels of serum leptin, cholesterol, LDL-C, and HDL-C along with APO E polymorphism were studied in 39 subjects with probable AD and 42 cognitive normal individuals. Results: AD group showed significantly lower levels of leptin (P = 0.00) as compared to control group. However, there was no significant difference in cholesterol, triglycerides, LDL-C, and HDL-C levels in AD and control groups. The frequency of ε4 allele in AD (38.5%) was found to be significantly higher than in control (10.3%). ε3 allele was more frequent than ε4 allele in AD and control group.
Collapse
Affiliation(s)
- Rachna Agarwal
- Department of Neurochemistry, Institute of Human Behaviour and Allied Sciences, Delhi, India
| | - Puneet Talwar
- Genetics and Molecular Medicine, Institute of Genomics and Integrated Biology, Delhi, India
| | - Suman S Kushwaha
- Department of Neurology, Institute of Human Behaviour and Allied Sciences, Delhi, India
| | | | - Ritushree Kukreti
- Genetics and Molecular Medicine, Institute of Genomics and Integrated Biology, Delhi, India
| |
Collapse
|
14
|
Chmyrov V, Spielmann T, Hevekerl H, Widengren J. Trans–Cis Isomerization of Lipophilic Dyes Probing Membrane Microviscosity in Biological Membranes and in Live Cells. Anal Chem 2015; 87:5690-7. [DOI: 10.1021/acs.analchem.5b00863] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Volodymyr Chmyrov
- Experimental
Biomolecular
Physics, Department of Applied Physics, Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Thiemo Spielmann
- Experimental
Biomolecular
Physics, Department of Applied Physics, Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Heike Hevekerl
- Experimental
Biomolecular
Physics, Department of Applied Physics, Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Jerker Widengren
- Experimental
Biomolecular
Physics, Department of Applied Physics, Royal Institute of Technology, Stockholm 106 91, Sweden
| |
Collapse
|
15
|
Lipids in Amyloid-β Processing, Aggregation, and Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 855:67-94. [PMID: 26149926 DOI: 10.1007/978-3-319-17344-3_3] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aggregation of amyloid-beta (Aβ) peptide is the major event underlying neuronal damage in Alzheimer's disease (AD). Specific lipids and their homeostasis play important roles in this and other neurodegenerative disorders. The complex interplay between the lipids and the generation, clearance or deposition of Aβ has been intensively investigated and is reviewed in this chapter. Membrane lipids can have an important influence on the biogenesis of Aβ from its precursor protein. In particular, increased cholesterol in the plasma membrane augments Aβ generation and shows a strong positive correlation with AD progression. Furthermore, apolipoprotein E, which transports cholesterol in the cerebrospinal fluid and is known to interact with Aβ or compete with it for the lipoprotein receptor binding, significantly influences Aβ clearance in an isoform-specific manner and is the major genetic risk factor for AD. Aβ is an amphiphilic peptide that interacts with various lipids, proteins and their assemblies, which can lead to variation in Aβ aggregation in vitro and in vivo. Upon interaction with the lipid raft components, such as cholesterol, gangliosides and phospholipids, Aβ can aggregate on the cell membrane and thereby disrupt it, perhaps by forming channel-like pores. This leads to perturbed cellular calcium homeostasis, suggesting that Aβ-lipid interactions at the cell membrane probably trigger the neurotoxic cascade in AD. Here, we overview the roles of specific lipids, lipid assemblies and apolipoprotein E in Aβ processing, clearance and aggregation, and discuss the contribution of these factors to the neurotoxicity in AD.
Collapse
|
16
|
Di Scala C, Chahinian H, Yahi N, Garmy N, Fantini J. Interaction of Alzheimer's β-amyloid peptides with cholesterol: mechanistic insights into amyloid pore formation. Biochemistry 2014; 53:4489-502. [PMID: 25000142 DOI: 10.1021/bi500373k] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Brain cholesterol plays a critical role in Alzheimer's disease and other neurodegenerative diseases. The molecular mechanisms linking cholesterol to neurotoxicity have remained elusive for a long time, but recent data have allowed the identification of functional cholesterol-binding domains in several amyloidogenic proteins involved in neurodegenerative diseases, including Alzheimer's disease. In this review, we analyze the cholesterol binding properties of β-amyloid (Aβ) peptides and the impact of these interactions on amyloid pore formation. We show that although the cholesterol-binding domains of Aβ peptides and of transmembrane precursor C99 are partially overlapping, they involve distinct amino acid residues, so that cholesterol has a greater affinity for Aβ than for C99. Synthetic 22-35 and 25-35 fragments of Aβ retained the ability of the full-length peptide 1-42 to bind cholesterol and to form zinc-sensitive, calcium-permeable amyloid pores in cultured neural cells. Studies with mutant peptides allowed the identification of key residues involved in cholesterol binding and channel formation. Cholesterol promoted the insertion of Aβ in the plasma membrane, induced α-helical structuration, and forced the peptide to adopt a tilted topology that favored the oligomerization process. Bexarotene, an amphipathic drug currently considered as a potential candidate medication for the treatment of neurodegenerative diseases, competed with cholesterol for binding to Aβ and prevented oligomeric channel formation. These studies indicate that it is possible to prevent the generation of neurotoxic oligomers by targeting the cholesterol-binding domain of Aβ peptides. This original strategy could be used for the treatment of Alzheimer's and other neurodegenerative diseases that involve cholesterol-dependent toxic oligomers.
Collapse
Affiliation(s)
- Coralie Di Scala
- EA-4674, Faculté des Sciences de Saint-Jérôme, Aix-Marseille Université , 13013 Marseille, France
| | | | | | | | | |
Collapse
|
17
|
Samtani MN, Raghavan N, Shi Y, Novak G, Farnum M, Lobanov V, Schultz T, Yang E, DiBernardo A, Narayan VA. Disease progression model in subjects with mild cognitive impairment from the Alzheimer's disease neuroimaging initiative: CSF biomarkers predict population subtypes. Br J Clin Pharmacol 2013; 75:146-61. [PMID: 22534009 DOI: 10.1111/j.1365-2125.2012.04308.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
AIM The objective is to develop a semi-mechanistic disease progression model for mild cognitive impairment (MCI) subjects. The model aims to describe the longitudinal progression of ADAS-cog scores from the Alzheimer's disease neuroimaging initiative trial that had data from 198 MCI subjects with cerebrospinal fluid (CSF) information who were followed for 3 years. METHOD Various covariates were tested on disease progression parameters and these variables fell into six categories: imaging volumetrics, biochemical, genetic, demographic, cognitive tests and CSF biomarkers. RESULTS CSF biomarkers were associated with both baseline disease score and disease progression rate in subjects with MCI. Baseline disease score was also correlated with atrophy measured using hippocampal volume. Progression rate was also predicted by executive functioning as measured by the Trail B-test. CONCLUSION CSF biomarkers have the ability to discriminate MCI subjects into sub-populations that exhibit markedly different rates of disease progression on the ADAS-cog scale. These biomarkers can therefore be utilized for designing clinical trials enriched with subjects that carry the underlying disease pathology.
Collapse
Affiliation(s)
- Mahesh N Samtani
- Johnson & Johnson Pharmaceutical Research & Development, Clinical Pharmacology Department, Raritan, New Jersey 08869, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Golfetto O, Hinde E, Gratton E. Laurdan fluorescence lifetime discriminates cholesterol content from changes in fluidity in living cell membranes. Biophys J 2013; 104:1238-47. [PMID: 23528083 DOI: 10.1016/j.bpj.2012.12.057] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 12/16/2012] [Accepted: 12/21/2012] [Indexed: 01/17/2023] Open
Abstract
Detection of the fluorescent properties of Laurdan has been proven to be an efficient tool to investigate membrane packing and ordered lipid phases in model membranes and living cells. Traditionally the spectral shift of Laurdan's emission from blue in the ordered lipid phase of the membrane (more rigid) toward green in the disordered lipid phase (more fluid) is quantified by the generalized polarization function. Here, we investigate the fluorescence lifetime of Laurdan at two different emission wavelengths and find that when the dipolar relaxation of Laurdan's emission is spectrally isolated, analysis of the fluorescence decay can distinguish changes in membrane fluidity from changes in cholesterol content. Using the phasor representation to analyze changes in Laurdan's fluorescence lifetime we obtain two different phasor trajectories for changes in polarity versus changes in cholesterol content. This gives us the ability to resolve in vivo membranes with different properties such as water content and cholesterol content and thus perform a more comprehensive analysis of cell membrane heterogeneity. We demonstrate this analysis in NIH3T3 cells using Laurdan as a biosensor to monitor changes in the membrane water content during cell migration.
Collapse
Affiliation(s)
- Ottavia Golfetto
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | | | | |
Collapse
|
19
|
Singh P, Saxena R, Srinivas G, Pande G, Chattopadhyay A. Cholesterol biosynthesis and homeostasis in regulation of the cell cycle. PLoS One 2013; 8:e58833. [PMID: 23554937 PMCID: PMC3598952 DOI: 10.1371/journal.pone.0058833] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 02/06/2013] [Indexed: 02/07/2023] Open
Abstract
The cell cycle is a ubiquitous, multi-step process that is essential for growth and proliferation of cells. The role of membrane lipids in cell cycle regulation is not explored well, although a large number of cytoplasmic and nuclear regulators have been identified. We focus in this work on the role of membrane cholesterol in cell cycle regulation. In particular, we have explored the stringency of the requirement of cholesterol in the regulation of cell cycle progression. For this purpose, we utilized distal and proximal inhibitors of cholesterol biosynthesis, and monitored their effect on cell cycle progression. We show that cholesterol content increases in S phase and inhibition of cholesterol biosynthesis results in cell cycle arrest in G1 phase under certain conditions. Interestingly, G1 arrest mediated by cholesterol biosynthesis inhibitors could be reversed upon metabolic replenishment of cholesterol. Importantly, our results show that the requirement of cholesterol for G1 to S transition is absolute, and even immediate biosynthetic precursors of cholesterol, differing with cholesterol merely in a double bond, could not replace cholesterol for reversing the cell cycle arrest. These results are useful in the context of diseases, such as cancer and Alzheimer’s disease, that are associated with impaired cholesterol biosynthesis and homeostasis.
Collapse
Affiliation(s)
- Pushpendra Singh
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
| | - Roopali Saxena
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
| | - Gunda Srinivas
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
| | - Gopal Pande
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
- * E-mail: (AC); (GP)
| | - Amitabha Chattopadhyay
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
- * E-mail: (AC); (GP)
| |
Collapse
|
20
|
Evidence for premature lipid raft aging in APP/PS1 double-transgenic mice, a model of familial Alzheimer disease. J Neuropathol Exp Neurol 2012; 71:868-81. [PMID: 22975585 DOI: 10.1097/nen.0b013e31826be03c] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Altered lipid raft homeostasis has been considered to contribute to cellular deregulation, leading to neuronal loss in Alzheimer disease. Alterations in these microdomains affect amyloid precursor protein (APP) processing, resulting in neurotoxic conditions, but modifications of the molecular structure of lipid rafts in Alzheimer disease model mice have not been characterized. Using a lipidomic approach, we investigated frontal cortex lipid rafts inwild-type mice and in double-transgenic APP/presenilin 1 (PS1) mice. Lipid rafts in wild-type mice undergo age-dependent modifications, that is, decreased cholesterol and sterol esters levels, augmented sphingomyelin and saturated fatty acid content, and increased phospholipids/cholesterol ratio. These age-dependent changes were more dramatic and occurred earlier in APP/PS1 mice; other lipid classes (e.g. sulfatides) and essential long-chain polyunsaturated fatty acids (including docosahexaenoic and arachidonic acids) were also affected. Steady state anisotropy measurements demonstrated that APP/PS1 animals exhibit more viscous (membrane-ordered) lipid rafts and that this is mainly attributable to reduced unsaturation of phospholipids and increased sphingomyelin levels rather than to changes in cholesterol. In summary, we demonstrate that aging is accompanied by alteration of the physicochemical structure of lipid raft microdomains. This "lipid raft aging," a metaphenomenon, is considerably exacerbated by the induced amyloid burden in APP/PS1 genotype.
Collapse
|
21
|
Meratan AA, Nemat-Gorgani M. Mitochondrial membrane permeabilization upon interaction with lysozyme fibrillation products: Role of mitochondrial heterogeneity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2149-57. [DOI: 10.1016/j.bbamem.2012.04.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 04/18/2012] [Accepted: 04/26/2012] [Indexed: 11/30/2022]
|
22
|
Ifuku M, Katafuchi T, Mawatari S, Noda M, Miake K, Sugiyama M, Fujino T. Anti-inflammatory/anti-amyloidogenic effects of plasmalogens in lipopolysaccharide-induced neuroinflammation in adult mice. J Neuroinflammation 2012; 9:197. [PMID: 22889165 PMCID: PMC3444880 DOI: 10.1186/1742-2094-9-197] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/25/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation involves the activation of glial cells in neurodegenerative diseases such as Alzheimer's disease (AD). Plasmalogens (Pls) are glycerophospholipids constituting cellular membranes and play significant roles in membrane fluidity and cellular processes such as vesicular fusion and signal transduction. METHODS In this study the preventive effects of Pls on systemic lipopolysaccharide (LPS)-induced neuroinflammation were investigated using immunohistochemistry, real-time PCR methods and analysis of brain glycerophospholipid levels in adult mice. RESULTS Intraperitoneal (i.p.) injections of LPS (250 μg/kg) for seven days resulted in increases in the number of Iba-1-positive microglia and glial fibrillary acidic protein (GFAP)-positive astrocytes in the prefrontal cortex (PFC) and hippocampus accompanied by the enhanced expression of IL-1β and TNF-α mRNAs. In addition, β-amyloid (Aβ3-16)-positive neurons appeared in the PFC and hippocampus of LPS-injected animals. The co-administration of Pls (i.p., 20 mg/kg) after daily LPS injections significantly attenuated both the activation of glial cells and the accumulation of Aβ proteins. Finally, the amount of Pls in the PFC and hippocampus decreased following the LPS injections and this reduction was suppressed by co-treatment with Pls. CONCLUSIONS These findings suggest that Pls have anti-neuroinflammatory and anti-amyloidogenic effects, thereby indicating the preventive or therapeutic application of Pls against AD.
Collapse
Affiliation(s)
- Masataka Ifuku
- Department of Integrative Physiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Bettermann K, Arnold AM, Williamson J, Rapp S, Sink K, Toole JF, Carlson MC, Yasar S, Dekosky S, Burke GL. Statins, risk of dementia, and cognitive function: secondary analysis of the ginkgo evaluation of memory study. J Stroke Cerebrovasc Dis 2012; 21:436-44. [PMID: 21236699 PMCID: PMC3140577 DOI: 10.1016/j.jstrokecerebrovasdis.2010.11.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 10/26/2010] [Accepted: 11/07/2010] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Lipid-lowering medications (LLMs) and especially statin drugs can delay cognitive decline and dementia onset in individuals with and without mild cognitive impairment (MCI) at baseline. METHODS A longitudinal, observational study was conducted of 3069 cognitively healthy elderly patients (≥75 years of age) who were enrolled in the Ginkgo Evaluation of Memory Study. The primary outcome measure was the time to adjudicated all-cause dementia and Alzheimer dementia (AD). The secondary outcome measure was the change in global cognitive function over time measured by scores from the Modified Mini-Mental State Exam (3MSE) and the cognitive subscale of the AD Assessment Scale (ADAS-Cog). RESULTS Among participants without MCI at baseline, the current use of statins was consistently associated with a reduced risk of all-cause dementia (hazard ratio [HR], 0.79; 95% confidence interval [95% CI], 0.65-0.96; P = .021) and AD (HR, 0.57; 95% CI, 0.39-0.85; P = .005). In participants who initiated statin therapy, lipophilic statins tended to reduce dementia risk more than nonlipophilic agents. In contrast, there was no significant association between LLM use (including statins), dementia onset, or cognitive decline in individuals with baseline MCI. However, in individuals without MCI at baseline, there was a trend for a neuroprotective effect of statins on cognitive decline. CONCLUSIONS Statins may slow the rate of cognitive decline and delay the onset of AD and all-cause dementia in cognitively healthy elderly individuals, whereas individuals with MCI may not have comparable cognitive protection from these agents. However, the results from this observational study need to be interpreted with caution and will require confirmation by randomized clinical trials stratifying treatment groups based on MCI status at baseline.
Collapse
Affiliation(s)
- Kerstin Bettermann
- Department of Neurology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Katafuchi T, Ifuku M, Mawatari S, Noda M, Miake K, Sugiyama M, Fujino T. Effects of plasmalogens on systemic lipopolysaccharide-induced glial activation and β-amyloid accumulation in adult mice. Ann N Y Acad Sci 2012; 1262:85-92. [DOI: 10.1111/j.1749-6632.2012.06641.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Lestaevel P, Bensoussan H, Racine R, Airault F, Gourmelon P, Souidi M. Transcriptomic effects of depleted uranium on acetylcholine and cholesterol metabolisms in Alzheimer's disease model. C R Biol 2011; 334:85-90. [PMID: 21333939 DOI: 10.1016/j.crvi.2010.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 11/25/2010] [Accepted: 12/03/2010] [Indexed: 11/16/2022]
Abstract
Some heavy metals, or aluminium, could participate in the development of Alzheimer disease (AD). Depleted uranium (DU), another heavy metal, modulates the cholinergic system and the cholesterol metabolism in the brain of rats, but without neurological disorders. The aim of this study was to determine what happens in organisms exposed to DU that will/are developing the AD. This study was thus performed on a transgenic mouse model for human amyloid precursor protein (APP), the Tg2576 strain. The possible effects of DU through drinking water (20 mg/L) over an 8-month period were analyzed on acetylcholine and cholesterol metabolisms at gene level in the cerebral cortex. The mRNA levels of choline acetyl transferase (ChAT) vesicular acetylcholine transporter (VAChT) and ATP-binding cassette transporter A1 (ABC A1) decreased in control Tg2576 mice in comparison with wild-type mice (respectively -89%, -86% and -44%, p < 0.05). Chronic exposure of Tg2576 mice to DU increased mRNA levels of ChAT (+189%, p < 0.05), VAChT (+120%, p < 0.05) and ABC A1 (+52%, p < 0.05) compared to control Tg2576 mice. Overall, these modifications of acetylcholine and cholesterol metabolisms did not lead to increased disturbances that are specific of AD, suggesting that chronic DU exposure did not worsen the pathology in this experimental model.
Collapse
Affiliation(s)
- Philippe Lestaevel
- Direction de la radioprotection de l'Homme, service de radiobiologie et d'épidémiologie, laboratoire de radiotoxicologie expérimentale, institut de radioprotection et de sûreté nucléaire, Fontenay-aux-Roses, France.
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Alzheimer disease (AD) is the most common causes of neurodegenerative disorder in the elderly individuals. Clinically, patients initially present with short-term memory loss, subsequently followed by executive dysfunction, confusion, agitation, and behavioral disturbances. Three causative genes have been associated with autosomal dominant familial AD (APP, PSEN1, and PSEN2) and 1 genetic risk factor (APOEε4 allele). Identification of these genes has led to a number of animal models that have been useful to study the pathogenesis underlying AD. In this article, we provide an overview of the clinical and genetic features of AD.
Collapse
Affiliation(s)
- Lynn M. Bekris
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Chang-En Yu
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Thomas D. Bird
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
27
|
Boche D, Denham N, Holmes C, Nicoll JAR. Neuropathology after active Abeta42 immunotherapy: implications for Alzheimer's disease pathogenesis. Acta Neuropathol 2010; 120:369-84. [PMID: 20632020 DOI: 10.1007/s00401-010-0719-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/07/2010] [Accepted: 07/07/2010] [Indexed: 12/26/2022]
Abstract
The amyloid cascade hypothesis of Alzheimer's disease (AD) is testable: it implies that interference with Abeta aggregation and plaque formation may be therapeutically useful. Abeta42 immunisation of amyloid precursor protein (APP) transgenic mice prevented plaque formation and caused removal of existing plaques. The first clinical studies of Abeta immunisation in AD patients (AN1792, Elan Pharmaceuticals) were halted when some patients suffered side effects. Since our confirmation that Abeta immunisation can prompt plaque removal in human AD, we have performed a clinical and neuropathological follow up of AD patients in the initial Elan Abeta immunisation trial. In immunised AD patients, we found: a lower Abeta load, with evidence that plaques had been removed; a reduced tau load in neuronal processes, but not in cell bodies; and no evidence of a beneficial effect on synapses. There were pathological "side effects" including: increased microglial activation; increased cerebral amyloid angiopathy; and there is some evidence for increased soluble/oligomeric Abeta. A pathophysiological mechanism involving effects on the cerebral vasculature is proposed for the clinical side effects observed with some active and passive vaccine protocols. Our current knowledge of the effects of Abeta immunotherapy is based on functional information from the early clinical trials and a few post mortem cases. Several further clinical studies are underway using a variety of protocols and important clinical, imaging and neuropathological data will become available in the near future. The information obtained will be important in helping to understand the pathogenesis not only of AD but also of other neurodegenerative disorders associated with protein aggregation.
Collapse
|
28
|
Minami A, Furuto A, Uemura M. Dynamic compositional changes of detergent-resistant plasma membrane microdomains during plant cold acclimation. PLANT SIGNALING & BEHAVIOR 2010; 5:1115-8. [PMID: 20818179 PMCID: PMC3115080 DOI: 10.4161/psb.5.9.12478] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Plants increase their freezing tolerance upon exposure to low, non-freezing temperatures, which is known as cold acclimation. Cold acclimation results in a decrease in the proportion of sphingolipids in the plasma membrane in many plants including Arabidopsis thaliana. The decrease in sphingolipids has been considered to contribute to the increase in the cryostability of the plasma membrane through regulating membrane fluidity. Recently we have proposed a possibility of another important sphingolipid function associated with cold acclimation. In animal cells, it has been known that the plasma membrane contains microdomains due to the chanracteristics of sphingolipids and sterols, and the sphingolipid- and sterol-enriched microdomains are thought to function as platforms for cell signaling, membrane trafficking and pathogen response. In our research on characterization of microdomain-associated lipids and proteins in Arabidopsis, cold-acclimation-induced decrease in sphingolipids resulted in a decrease of microdomains in the plasma membrane and there were considerable changes in membrane transport-, cytoskeleton- and endocytosis-related proteins in the microdomains during cold acclimation. Based on these results, we discuss a functional relationship between the changes in microdomain components and plant cold acclimation.
Collapse
Affiliation(s)
- Anzu Minami
- Cryobiofrontier Research Center, Iwate University, Morioka, Japan
| | | | | |
Collapse
|
29
|
Molecular insights into amyloid regulation by membrane cholesterol and sphingolipids: common mechanisms in neurodegenerative diseases. Expert Rev Mol Med 2010; 12:e27. [PMID: 20807455 PMCID: PMC2931503 DOI: 10.1017/s1462399410001602] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer, Parkinson and other neurodegenerative diseases involve a series of brain
proteins, referred to as ‘amyloidogenic proteins’, with exceptional
conformational plasticity and a high propensity for self-aggregation. Although the
mechanisms by which amyloidogenic proteins kill neural cells are not fully understood, a
common feature is the concentration of unstructured amyloidogenic monomers on
bidimensional membrane lattices. Membrane-bound monomers undergo a series of
lipid-dependent conformational changes, leading to the formation of oligomers of varying
toxicity rich in β-sheet structures (annular pores, amyloid fibrils) or in
α-helix structures (transmembrane channels). Condensed membrane nano- or
microdomains formed by sphingolipids and cholesterol are privileged sites for the binding
and oligomerisation of amyloidogenic proteins. By controlling the balance between
unstructured monomers and α or β conformers (the chaperone effect),
sphingolipids can either inhibit or stimulate the oligomerisation of amyloidogenic
proteins. Cholesterol has a dual role: regulation of protein–sphingolipid
interactions through a fine tuning of sphingolipid conformation (indirect effect), and
facilitation of pore (or channel) formation through direct binding to amyloidogenic
proteins. Deciphering this complex network of molecular interactions in the context of
age- and disease-related evolution of brain lipid expression will help understanding of
how amyloidogenic proteins induce neural toxicity and will stimulate the development of
innovative therapies for neurodegenerative diseases.
Collapse
|
30
|
Siu H, Duhamel J, Sasaki DY, Pincus JL. Nanodomain formation in lipid membranes probed by time-resolved fluorescence. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:10985-10994. [PMID: 20536249 DOI: 10.1021/la9045429] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Time-resolved fluorescence measurements on liposomes prepared with 1 mol % pyrene-labeled lipids (PLLs) with a headgroup bearing either an alcohol (PSOH) or an imido diacetic acid (PSIDA) and 99 mol % 1-palmitoyl-2-oleyl-3-sn-phosphatidylcholines (POPC) or 99 mol % distearylphosphatidylcholine (DSPC) were performed to investigate how lipids phase separate within the membrane bilayer. Global analysis of the fluorescence decays with the fluorescence blob model (FBM) led to the conclusion that the PLLs were homogeneously distributed on the surface of POPC vesicles while the PLLs phase-separated in the DSPC vesicles. The analysis yielded the fraction of aggregated pyrenes, f(agg). The large f(agg) values found for PSIDA suggest that the imido diacetic acid headgroup of PSIDA induces self-aggregation and phase separation in both membranes. The addition of external cations such as Cu(2+) and La(3+) was shown to hinder diffusional encounters between PSIDAs. The cations seem to target preferentially unassociated PSIDAs rather than aggregated PSIDA clusters. Accounting for the quenching of pyrene by Cu(2+) enables one to use PSIDA to probe the microviscosity of the lipid membrane. Using this effect, the environment of PSIDA in the DSPC membrane was found to be about 6 times more viscous than that in the POPC membrane. This difference is attributed to the difference in viscosity of the fluid POPC membrane and the gel-like DSPC membranes.
Collapse
Affiliation(s)
- Howard Siu
- Institute for Polymer Research, Department of Chemistry, University of Waterloo, Waterloo ON N2L 3G1, Canada
| | | | | | | |
Collapse
|
31
|
Weber P, Wagner M, Schneckenburger H. Fluorescence imaging of membrane dynamics in living cells. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:046017. [PMID: 20799819 DOI: 10.1117/1.3470446] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Methods of wide-field fluorescence microscopy for measuring membrane dynamics of living cells are described, including spectral imaging as well as anisotropy imaging of the membrane marker 6-dodecanoyl-2-dimethylamino naphthalene (laurdan). Plasma membranes are selected by illumination with an evanescent electromagnetic field and distinguished from intracellular membranes assessed by whole-cell illumination. While fluorescence spectra of laurdan appeared red-shifted with decreasing membrane stiffness, fluorescence anisotropy and rotational correlation times were reduced with increasing membrane fluidity. Membrane stiffness was found to increase with decreasing temperature and increasing amounts of cholesterol and was always higher for the plasma membrane than for intracellular membranes. These effects may have some clinical relevance in the research of drug resistance or cell aging.
Collapse
Affiliation(s)
- Petra Weber
- Hochschule Aalen, Institut fur Angewandte Forschung, Aalen, Germany
| | | | | |
Collapse
|
32
|
Kumari U, Heese K. Cardiovascular dementia - a different perspective. Open Biochem J 2010; 4:29-52. [PMID: 20448820 PMCID: PMC2864432 DOI: 10.2174/1874091x01004010029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 02/08/2023] Open
Abstract
The number of dementia patients has been growing in recent years and dementia represents a significant threat to aging people all over the world. Recent research has shown that the number of people affected by Alzheimer's disease (AD) and dementia is growing at an epidemic pace. The rapidly increasing financial and personal costs will affect the world's economies, health care systems, and many families. Researchers are now exploring a possible connection among AD, vascular dementia (VD), diabetes mellitus (type 2, T2DM) and cardiovascular diseases (CD). This correlation may be due to a strong association of cardiovascular risk factors with AD and VD, suggesting that these diseases share some biologic pathways. Since heart failure is associated with an increased risk of AD and VD, keeping the heart healthy may prove to keep the brain healthy as well. The risk for dementia is especially high when diabetes mellitus is comorbid with severe systolic hypertension or heart disease. In addition, the degree of coronary artery disease (CAD) is independently associated with cardinal neuropathological lesions of AD. Thus, the contribution of T2DM and CD to AD and VD implies that cardiovascular therapies may prove useful in preventing AD and dementia.
Collapse
Affiliation(s)
- Udhaya Kumari
- Division of Cell and Molecular Biology, School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | | |
Collapse
|
33
|
Abstract
In Alzheimer's disease (AD), there is abnormal accumulation of Abeta and tau proteins in the brain. There is an associated immunological response, but it is still unclear whether this is beneficial or harmful. Inflammation in AD, specifically in the form of microglial activation, has, for many years, been considered to contribute to disease progression. However, two types of evidence suggest that it may be appropriate to revise this view: first, the disappointing results of prospective clinical trials of anti-inflammatory agents and, second, the observation that microglia can clear plaques in AD following Abeta immunization. Although Abeta immunization alters AD pathology, there is limited evidence so far of benefit to cognitive function. Immunization against microorganisms is almost always used as a method of disease prevention rather than to treat a disease process that has already started. In animal models, immunotherapy at an early age can protect against Abeta accumulation and it will be interesting to see if this can usefully be applied to humans to prevent AD.
Collapse
Affiliation(s)
- Delphine Boche
- Division of Clinical Neurosciences, University of Southampton, Southampton, UK.
| | | |
Collapse
|
34
|
Paul V, Meyer HHD, Leidl K, Soumian S, Albrecht C. A novel enzyme immunoassay specific for ABCA1 protein quantification in human tissues and cells. J Lipid Res 2008; 49:2259-67. [PMID: 18541924 DOI: 10.1194/jlr.d700040-jlr200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) mediates the transport of cholesterol and phospholipids from cells to lipid-poor HDL and maintains cellular lipid homeostasis. Impaired ABCA1 function plays a role in lipid disorders, cardiovascular disease, atherosclerosis, and metabolic disorders. Despite the clinical importance of ABCA1, no method is available for quantifying ABCA1 protein. We developed a sensitive indirect competitive ELISA for measuring ABCA1 protein in human tissues using a commercial ABCA1 peptide and a polyclonal anti-ABCA1 antibody. The ELISA has a detection limit of 8 ng/well (0.08 mg/l) with a working range of 9-1000 ng/well (0.09-10 mg/l). Intra- and interassay coefficient of variations (CVs) were 6.4% and 9.6%, respectively. Good linearity (r = 0.97-0.99) was recorded in serial dilutions of human arterial and placental crude membrane preparations, and fibroblast lysates. The ELISA measurements for ABCA1 quantification in reference arterial tissues corresponded well with immunoblot analysis. The assay performance and clinical utility was evaluated with arterial tissues obtained from 15 controls and 44 patients with atherosclerotic plaques. ABCA1 protein concentrations in tissue lysates were significantly lower in patients (n = 24) as compared with controls (n = 5; 9.37 +/- 0.82 vs. 17.03 +/- 4.25 microg/g tissue; P < 0.01). The novel ELISA enables the quantification of ABCA1 protein in human tissues and confirms previous semiquantitative immunoblot results.
Collapse
Affiliation(s)
- Vijay Paul
- Physiology Weihenstephan, Technical University Munich, Weihenstephaner Berg 3, 85350 Freising, Germany
| | | | | | | | | |
Collapse
|
35
|
Gamerdinger M, Clement AB, Behl C. Effects of sulindac sulfide on the membrane architecture and the activity of gamma-secretase. Neuropharmacology 2008; 54:998-1005. [PMID: 18359496 DOI: 10.1016/j.neuropharm.2008.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2007] [Revised: 01/22/2008] [Accepted: 02/08/2008] [Indexed: 11/28/2022]
Abstract
gamma-Secretase is a membrane-embedded multi-protein complex that catalyzes the final cut of the Alzheimer's disease-related amyloid precursor protein (APP) to amyloid-beta peptides of variable length (37-43 amino acids) via an unusual intramembrane cleavage. Recent findings propose that some commonly used non-steroidal anti-inflammatory drugs (NSAIDs) have the ability to modulate specifically gamma-secretase activity without inhibiting the enzyme as a whole. These drugs may shift the processing of APP from the longer amyloid-beta 42 peptide towards shorter, less fibrillogenic and less toxic amyloid-beta species. We hypothesize that gamma-secretase activity, as an enzyme that is strictly associated with cellular membranes, is sensitive to alterations of the hydrophobic membrane environment. Here, we show that the gamma-secretase modulator and amyloid-beta 42-lowering drug sulindac sulfide alters the physical state of the membrane and strongly decreases fluidity of cellular membranes. Furthermore, sulindac sulfide changed the protein composition of membrane microdomains, the so-called lipid rafts. Most significantly, APP C-terminal fragments (CTFs) were redistributed from rafts towards non-raft membrane domains. This could be demonstrated also in cell-free assays, where in addition presenilin-1, the catalytic subunit of the gamma-secretase complex, was shifted out of lipid rafts. Together, these findings suggest that sulindac sulfide directly alters the membrane architecture and shifts the gamma-secretase-mediated cleavage of APP towards a hydrophobic environment where the enzyme-substrate complex is in a conformation for processing preferentially shorter amyloid-beta peptides.
Collapse
Affiliation(s)
- Martin Gamerdinger
- Department of Pathobiochemistry, Institute for Physiological Chemistry and Pathobiochemistry, Medical School, Johannes Gutenberg University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | | | | |
Collapse
|
36
|
Won JS, Im YB, Khan M, Contreras M, Singh AK, Singh I. Lovastatin inhibits amyloid precursor protein (APP) beta-cleavage through reduction of APP distribution in Lubrol WX extractable low density lipid rafts. J Neurochem 2008; 105:1536-49. [PMID: 18266936 DOI: 10.1111/j.1471-4159.2008.05283.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Previous studies have described that statins (inhibitors of cholesterol and isoprenoid biosynthesis) inhibit the output of amyloid-beta (Abeta) in the animal model and thus decrease risk of Alzheimer's disease. However, their action mechanism(s) in Abeta precursor protein (APP) processing and Abeta generation is not fully understood. In this study, we report that lovastatin treatment reduced Abeta output in cultured hippocampal neurons as a result of reduced APP levels and beta-secretase activities in low density Lubrol WX (non-ionic detergent) extractable lipid rafts (LDLR). Rather than altering cholesterol levels in lipid raft fractions and thus disrupting lipid raft structure, lovastatin decreased Abeta generation through down-regulating geranylgeranyl-pyrophosphate dependent endocytosis pathway. The inhibition of APP endocytosis by treatment with lovastatin and reduction of APP levels in LDLR fractions by treatment with phenylarsine oxide (a general endocytosis inhibitor) support the involvement of APP endocytosis in APP distribution in LDLR fractions and subsequent APP beta-cleavage. Moreover, lovastatin-mediated down-regulation of endocytosis regulators, such as early endosomal antigen 1, dynamin-1, and phosphatidylinositol 3-kinase activity, indicates that lovastatin modulates APP endocytosis possibly through its pleiotropic effects on endocytic regulators. Collectively, these data report that lovastatin mediates inhibition of LDLR distribution and beta-cleavage of APP in a geranylgeranyl-pyrophosphate and endocytosis-dependent manner.
Collapse
Affiliation(s)
- Je-Seong Won
- Department of Pathology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
37
|
Vestergaard M, Hamada T, Takagi M. Using model membranes for the study of amyloid beta:lipid interactions and neurotoxicity. Biotechnol Bioeng 2008; 99:753-63. [DOI: 10.1002/bit.21731] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
38
|
Abstract
Pharmacological treatment in Alzheimer's disease (AD) accounts for 10-20% of direct costs, and fewer than 20% of AD patients are moderate responders to conventional drugs (donepezil, rivastigmine, galantamine, memantine), with doubtful cost-effectiveness. Both AD pathogenesis and drug metabolism are genetically regulated complex traits in which hundreds of genes cooperatively participate. Structural genomics studies demonstrated that more than 200 genes might be involved in AD pathogenesis regulating dysfunctional genetic networks leading to premature neuronal death. The AD population exhibits a higher genetic variation rate than the control population, with absolute and relative genetic variations of 40-60% and 0.85-1.89%, respectively. AD patients also differ in their genomic architecture from patients with other forms of dementia. Functional genomics studies in AD revealed that age of onset, brain atrophy, cerebrovascular hemodynamics, brain bioelectrical activity, cognitive decline, apoptosis, immune function, lipid metabolism dyshomeostasis, and amyloid deposition are associated with AD-related genes. Pioneering pharmacogenomics studies also demonstrated that the therapeutic response in AD is genotype-specific, with apolipoprotein E (APOE) 4/4 carriers the worst responders to conventional treatments. About 10-20% of Caucasians are carriers of defective cytochrome P450 (CYP) 2D6 polymorphic variants that alter the metabolism and effects of AD drugs and many psychotropic agents currently administered to patients with dementia. There is a moderate accumulation of AD-related genetic variants of risk in CYP2D6 poor metabolizers (PMs) and ultrarapid metabolizers (UMs), who are the worst responders to conventional drugs. The association of the APOE-4 allele with specific genetic variants of other genes (e.g., CYP2D6, angiotensin-converting enzyme [ACE]) negatively modulates the therapeutic response to multifactorial treatments affecting cognition, mood, and behavior. Pharmacogenetic and pharmacogenomic factors may account for 60-90% of drug variability in drug disposition and pharmacodynamics. The incorporation of pharmacogenetic/pharmacogenomic protocols to AD research and clinical practice can foster therapeutics optimization by helping to develop cost-effective pharmaceuticals and improving drug efficacy and safety.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute for CNS Disorders, Bergondo, Coruña, Spain
| |
Collapse
|
39
|
Goodenowe DB, Cook LL, Liu J, Lu Y, Jayasinghe DA, Ahiahonu PWK, Heath D, Yamazaki Y, Flax J, Krenitsky KF, Sparks DL, Lerner A, Friedland RP, Kudo T, Kamino K, Morihara T, Takeda M, Wood PL. Peripheral ethanolamine plasmalogen deficiency: a logical causative factor in Alzheimer's disease and dementia. J Lipid Res 2007; 48:2485-98. [PMID: 17664527 DOI: 10.1194/jlr.p700023-jlr200] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although dementia of the Alzheimer's type (DAT) is the most common form of dementia, the severity of dementia is only weakly correlated with DAT pathology. In contrast, postmortem measurements of cholinergic function and membrane ethanolamine plasmalogen (PlsEtn) content in the cortex and hippocampus correlate with the severity of dementia in DAT. Currently, the largest risk factor for DAT is age. Because the synthesis of PlsEtn occurs via a single nonredundant peroxisomal pathway that has been shown to decrease with age and PlsEtn is decreased in the DAT brain, we investigated potential relationships between serum PlsEtn levels, dementia severity, and DAT pathology. In total, serum PlsEtn levels were measured in five independent population collections comprising >400 clinically demented and >350 nondemented subjects. Circulating PlsEtn levels were observed to be significantly decreased in serum from clinically and pathologically diagnosed DAT subjects at all stages of dementia, and the severity of this decrease correlated with the severity of dementia. Furthermore, a linear regression model predicted that serum PlsEtn levels decrease years before clinical symptoms. The putative roles that PlsEtn biochemistry play in the etiology of cholinergic degeneration, amyloid accumulation, and dementia are discussed.
Collapse
|
40
|
Germano M, Meleleo D, Montorfano G, Adorni L, Negroni M, Berra B, Rizzo AM. Plasma, red blood cells phospholipids and clinical evaluation after long chain omega-3 supplementation in children with attention deficit hyperactivity disorder (ADHD). Nutr Neurosci 2007; 10:1-9. [PMID: 17539477 DOI: 10.1080/10284150601153801] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Omega-3 and omega-6 long-chain polyunsaturated fatty acids (LCPUFAs), are crucial to brain development and function. Increasing evidence indicates that deficiencies or metabolic imbalances of these fatty acids might be associated with childhood developmental and psychiatric disorders including attention-deficit/hyperactivity disorder (ADHD). Omega-3 are often lacking on modern diets. Moreover preliminary evidences suggest that supplementation with omega-3 LCPUFAs, might help in the management of the ADHD linked behavioural and learning difficulties. However, few studies published to date have involved different populations, study designs, treatments and outcome results. Thus, further researches are required to assess the durability of the treatment effects, to determine optimal composition and dosages of the supplement and to develop reliable ways to identify patients that might have some benefits from this kind of treatment, also because the study of LCPUFAs and their metabolism might offer new approaches to the early identification and management of ADHD. In this paper, we provide new insight on the lipid pattern in plasma and red blood cells (RBC) phospholipids, together with evaluation of the arachidonic acid (AA)/eicosapentaenoic acid (EPA) ratio which seems to correlate with the improvement of the patients both from a biochemical and clinical point of view.
Collapse
Affiliation(s)
- Michele Germano
- NPI L R.C.S. Casa Sollievo della Sofferenza S. Giovanni R. (FG), Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Kristofiková Z, Kopecký V, Hofbauerová K, Hovorková P, Rípová D. Complex of Amyloid β Peptides with 24-Hydroxycholesterol and Its Effect on Hemicholinium-3 Sensitive Carriers. Neurochem Res 2007; 33:412-21. [PMID: 17717740 DOI: 10.1007/s11064-007-9443-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Accepted: 07/12/2007] [Indexed: 01/07/2023]
Abstract
Brains of Alzheimer disease patients in early stages of dementia contain an increased 24(S)-hydroxycholesterol (cerebrosterol)/cholesterol ratio when compared to controls. In this study, effects of amyloid beta peptides and of racemic 24-hydroxycholesterol were evaluated in vitro on undepleted or cholesterol-depleted hippocampal synaptosomes of young and old rats via a high-affinity choline transport and membrane anisotropy measurements. Depletion of membrane cholesterol decreased the transport of [3H]choline, increased the specific binding of [3H]hemicholinium-3 and decreased membrane anisotropy. However, less alterations were found in old when compared to young brains. 500 nM nonaggregated peptides were ineffective but aggregated fragment 1-42 evoked marked drops in the transport and anisotropy values on depleted synaptosomes. 50 microM 24-hydroxycholesterol inhibited choline transport on depleted synaptosomes but it did not influence membrane anisotropy. Peptides eliminated the actions of oxysterol on choline carriers in young but not in old rats. On the other hand, oxysterol eliminated the effects of peptides on membrane anisotropy. Our study suggests a possible role of membrane cholesterol in the regulation of choline carriers and supports data reporting a protective role of membrane cholesterol against toxic effects of amyloid beta peptides. Moreover, via Raman spectroscopy we demonstrate for the first time that peptides form a complex with 24-hydroxycholesterol.
Collapse
Affiliation(s)
- Zdena Kristofiková
- Alzheimer Disease Centre, Prague Psychiatric Centre, Ustavní 91, Prague 8 - Bohnice 181 03, Czech Republic.
| | | | | | | | | |
Collapse
|
42
|
Famer D, Crisby M. Rosuvastatin reduces gliosis and the accelerated weight gain observed in WT and ApoE−/− mice exposed to a high cholesterol diet. Neurosci Lett 2007; 419:68-73. [PMID: 17433542 DOI: 10.1016/j.neulet.2007.03.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 03/15/2007] [Accepted: 03/27/2007] [Indexed: 10/23/2022]
Abstract
The influence of a high cholesterol (HC) diet on brain pathology is being recognized increasingly and is of immense interest. Previous findings from our laboratory demonstrated that a high cholesterol diet increases gliosis, astrocytic reactivity and neuroinflammation in both wild type (WT) and apolipoprotein knockout (ApoE-/-) mice. In the present study, we analyzed whether this increase in astrocytic reactivity, monitored by the number of cells in the hippocampus labelled with glial fibrillary acidic protein (GFAP), could be reduced by the use of rosuvastatin, a potent competitive inhibitor of 3-hydroxy-3-methylglutaryl-Coenzyme A (HMG-CoA) reductase. Furthermore, we studied the effect of rosuvastatin on changes in lipoprotein levels and weight gain, and their correlation to gliosis, in mice fed a high cholesterol diet. A significant increase in weight, total-cholesterol (TC) and low-density lipoprotein (LDL) levels were observed in WT and ApoE-/- mice on a HC diet. The number of GFAP labelled cells was found to be significantly increased in mice on a HC diet and reduced in rosuvastatin-treated WT and ApoE-/- mice on a HC diet. A significant reduction of weight, total-cholesterol and LDL levels was observed in rosuvastatin-treated WTHC mice. Significant correlations were found between changes in body weight, GFAP labelled cells and plasma total-cholesterol levels in WT and ApoE-/- mice. However, the correlations were found to be weaker for the GFAP labelled cells in the ApoE-/- mice. The results indicate that the observed reduction of gliosis by rosuvastatin treatment may be due to mechanisms that are independent of its lipid-lowering effect.
Collapse
Affiliation(s)
- Daniel Famer
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden
| | | |
Collapse
|
43
|
Chauhan NB, Siegel GJ. Antisense inhibition at the beta-secretase-site of beta-amyloid precursor protein reduces cerebral amyloid and acetyl cholinesterase activity in Tg2576. Neuroscience 2007; 146:143-51. [PMID: 17303345 PMCID: PMC1955231 DOI: 10.1016/j.neuroscience.2007.01.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 12/27/2006] [Accepted: 01/05/2007] [Indexed: 12/29/2022]
Abstract
Misprocessing of beta-amyloid precursor protein (APP) leading to the formation of elevated quantities of beta-amyloid peptide (Abeta), derived by a cleavage at the beta-secretase site (N-671/673aa) and by a cleavage at the gamma-secretase site (C-711/713aa) of APP, is considered a key event in the pathogenesis of Alzheimer disease (AD). Point mutations near the beta-secretase site in the human gene for APP, such as in the Swedish mutation-KM670/671NL, lead to a form of dominantly inherited AD. These mutations are known to promote beta-site cleavage and to increase levels of Abeta. Abeta has been shown previously to increase acetyl cholinesterase (AChE) activity in vitro. We wished to test whether translational blocking of APP-mRNA at the mutated beta-site by antisense (AS) oligodeoxynucleotides (ODNs) directed to the mutated site will reduce cerebral amyloid in the Swedish transgenic mouse model (Tg2576). Mice were injected i.c.v. with AS-ODNs directed at the mutated beta-site (AS-beta site) or with AS-ODNs directed at the normal gamma-site (AS-gamma site) of human APP-mRNA, and compared with procedural controls that received i.c.v. injections of sense ODNs at the beta-site (S-beta site), sense ODNs at the gamma-site (S-gamma site) or mismatched ODNs, and with untreated littermates (Lt) and untreated transgenic mice (Tgs). ODNs were injected into the 3rd ventricle once a week for 4 weeks. Brains were processed for enzyme-linked immunosorbent assay analysis of beta- and gamma-cleaved soluble Abeta40 (sAbeta40), beta- and gamma-cleaved soluble Abeta42 (sAbeta42) and alpha-cleaved soluble beta-amyloid precursor protein (sAPPalpha). The physiological relevance of AS ODNs was tested by evaluating the cerebral distribution of AChE before and after the treatment. AChE was found increased about fivefold in Tg cortex as compared with control brain. Results show that compared with untreated and procedural controls, AS-beta increased cerebral levels of sAPPalpha by 43% and reduced sAbeta40/42 by approximately 39%; while simultaneously reducing the cortical density of AChE by approximately fourfold in the treated Tg animals, almost to the level found in the control brain (all values P<0.0001, analysis of variance, unpaired two-tailed Student's t-test), while AS-gamma did not have any effect. These results indicate that AS directed to the mutated beta-site may be an effective approach to treat familial AD.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Department of Anesthesiology, University of Illinois at Chicago 60612, and Neurology Service (127), Edward Hines, Jr., VA Hospital, Hines, IL 60614, USA.
| | | |
Collapse
|
44
|
Dodson SE, Gearing M, Lippa CF, Montine TJ, Levey AI, Lah JJ. LR11/SorLA expression is reduced in sporadic Alzheimer disease but not in familial Alzheimer disease. J Neuropathol Exp Neurol 2006; 65:866-72. [PMID: 16957580 PMCID: PMC2663339 DOI: 10.1097/01.jnen.0000228205.19915.20] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
LR11 is an ApoE receptor that is enriched in the brain. We have shown that LR11 is markedly downregulated in patients with sporadic Alzheimer disease (AD). This finding led us to explore whether reduced LR11 expression reflects a primary mechanism of disease or merely a secondary consequence of other AD-associated changes. Therefore, LR11 expression was assessed in a transgenic mouse model of AD and familial AD (FAD) brains. Immunohistochemistry and immunoblotting of LR11 in PS1/APP transgenic and wild-type mice indicated that LR11 levels are not affected by genotype or accumulation of amyloid pathology. LR11 expression was also evaluated based on immunoblotting and LR11 immunostaining intensity in human frontal cortex in controls, sporadic AD, and FAD, including cases with presenilin-1 (PS1) and presenilin-2 (PS2) mutations. Although LR11 was reduced in sporadic AD, there was no difference in protein level or staining intensity between control and FAD cases. The finding that LR11 expression is unaffected in both a mouse model of AD and autosomal-dominant forms of AD suggests that LR11 is not regulated by amyloid accumulation or other AD neuropathologic changes. We hypothesize that LR11 loss may be specific to sporadic AD and influence amyloid pathology through mechanisms independent of substrate-enzyme interactions regulated by FAD mutations.
Collapse
Affiliation(s)
- Sara E Dodson
- Center for Neurodegenerative Disease, Department of Neurology, Emory University Atlanta, Georgia 30322, USA
| | | | | | | | | | | |
Collapse
|
45
|
King SR, Smith AGA, Alpy F, Tomasetto C, Ginsberg SD, Lamb DJ. Characterization of the putative cholesterol transport protein metastatic lymph node 64 in the brain. Neuroscience 2006; 139:1031-8. [PMID: 16549269 DOI: 10.1016/j.neuroscience.2006.01.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Revised: 12/20/2005] [Accepted: 01/15/2006] [Indexed: 11/25/2022]
Abstract
Intracellular management of cholesterol is a critical process in the brain. Deficits with cholesterol transport and storage are linked to neurodegenerative disorders such as Neimann-Pick disease type C and Alzheimer's disease. One protein putatively involved in cholesterol transport is metastatic lymph node 64 (MLN64). MLN64 localizes to late endosomes which are part of the cholesterol internalization pathway. However, a detailed pattern of MLN64 expression in the brain is unclear. Using immunocytochemical and immunoblot analyses, we demonstrated the presence of MLN64 in several tissue types and various regions within the brain. MLN64 immunostaining in the CNS was heterogeneous, indicating selective expression in discrete specific cell populations and regions. MLN64 immunoreactivity was detected in glia and neurons, which displayed intracellular labeling consistent with an endosomal localization. Although previous studies suggested that MLN64 may promote steroid production in the brain, MLN64 immunoreactivity did not colocalize with steroidogenic cells in the CNS. These results demonstrate that MLN64 is produced in the mouse and human CNS in a restricted pattern of expression, suggesting that MLN64 serves a cell-specific function in cholesterol transport.
Collapse
Affiliation(s)
- S R King
- Scott Department of Urology, Room N730, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Escribá PV. Membrane-lipid therapy: a new approach in molecular medicine. Trends Mol Med 2006; 12:34-43. [PMID: 16325472 DOI: 10.1016/j.molmed.2005.11.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 10/27/2005] [Accepted: 11/17/2005] [Indexed: 10/25/2022]
Abstract
Although most drugs bind to proteins and regulate their activity, some drugs act through a new therapeutic approach called membrane-lipid therapy and bind to lipids, thus modulating the structure of membranes. Most cellular functions are highly dependent on the lipid environment because they are controlled by proteins in or around membranes. The wide variety of cell and organelle membranes and the existence of special lipid regions (e.g. microvilli) and domains (e.g. lipid rafts) support the possibility of designing specific lipid therapies. Indeed, recent evidence suggests that lipid therapy might have potential for the treatment of cancer, cardiovascular pathologies, neurodegenerative processes, obesity, metabolic disorders, inflammation, and infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Pablo V Escribá
- Molecular and Cellular Biomedicine, Associate Unit of the Consejo Superior de Investigaciones Científicas, IUNICS, Department of Biology, University of the Balearic Islands, E-07122 Palma de Mallorca, Spain.
| |
Collapse
|
47
|
Okuhira KI, Fitzgerald ML, Sarracino DA, Manning JJ, Bell SA, Goss JL, Freeman MW. Purification of ATP-binding Cassette Transporter A1 and Associated Binding Proteins Reveals the Importance of β1-Syntrophin in Cholesterol Efflux. J Biol Chem 2005; 280:39653-64. [PMID: 16192269 DOI: 10.1074/jbc.m510187200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) plays a critical role in HDL cholesterol metabolism, but the mechanism by which it transports lipid across membranes is poorly understood. Because growing evidence implicates accessory proteins in this process, we developed a method by which proteins interacting with the intact transporter could be identified. cDNAs encoding wild-type ABCA1 and a mutant lacking the C-terminal PDZ binding motif of ABCA1 were transfected into 293 cells, and the expressed proteins were solubilized using detergent conditions (0.75% CHAPS, 1 mg/ml phosphatidylcholine) predicted to retain high affinity protein-protein interactions. Proteins that co-purified with ABCA1 on an antibody affinity column were identified by liquid chromatographymass spectrometric analysis. A novel interaction with the PDZ protein beta1-syntrophin was identified using this approach, and this interaction was confirmed in human THP-1 macrophages and in mouse liver. Small interference RNA inhibition of beta1-syntrophin expression reduced cholesterol efflux from primary skin fibroblasts by 50% while decreasing efflux 30% in bone marrow-derived macrophages. Inhibition of beta1-syntrophin decreased ABCA1 protein levels, whereas overexpression of beta1-syntrophin increased ABCA1 cell-surface expression and stimulated efflux to apolipoprotein A-I. These findings indicate that beta1-syntrophin acts through a class-I PDZ interaction with the C terminus of ABCA1 to regulate the cellular distribution and activity of the transporter. The approach used to identify beta1-syntrophin as an ABCA1-binding protein should prove useful in elucidating other protein interactions upon which ABCA1 function depends.
Collapse
Affiliation(s)
- Kei-ichiro Okuhira
- Lipid Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Kretzschmar D. Neurodegenerative mutants in Drosophila: a means to identify genes and mechanisms involved in human diseases? INVERTEBRATE NEUROSCIENCE 2005; 5:97-109. [PMID: 16187075 DOI: 10.1007/s10158-005-0005-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 07/20/2005] [Indexed: 01/10/2023]
Abstract
There are 50 ways to leave your lover (Simon 1987) but many more to kill your brain cells. Several neurodegenerative diseases in humans, like Alzheimer's disease, have been intensely studied but the underlying cellular and molecular mechanisms are still unknown for most of them. For those syndromes where associated gene products have been identified their biochemistry and physiological as well as pathogenic function is often still under debate. This is in part due to the inherent limitations of genetic analyses in humans and other mammals and therefore experimentally accessible invertebrate in vivo models, such as Caenorhabditis elegans and Drosophila melanogaster, have recently been introduced to investigate neurodegenerative syndromes. Several laboratories have used transgenic approaches in Drosophila to study the human genes associated with neurodegenerative diseases. This has added substantially to our understanding of the mechanisms leading to neurodegenerative diseases in humans. The isolation and characterization of Drosophila mutants, which display a variety of neurodegenerative phenotypes, also provide valuable insights into genes, pathways, and mechanisms causing neurodegeneration. So far only about two dozen such mutants have been described but already their characterization reveals an involvement of various cellular functions in neurodegeneration, ranging from preventing oxidative stress to RNA editing. Some of the isolated genes can already be associated with human neurodegenerative diseases and hopefully the isolation and characterization of more of these mutants, together with an analysis of homologous genes in vertebrate models, will provide insights into the genetic and molecular basis of human neurodegenerative diseases.
Collapse
Affiliation(s)
- Doris Kretzschmar
- Center for Research on Occupational and Environmental Toxicology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97201, USA.
| |
Collapse
|
49
|
Vigh L, Escribá PV, Sonnleitner A, Sonnleitner M, Piotto S, Maresca B, Horváth I, Harwood JL. The significance of lipid composition for membrane activity: New concepts and ways of assessing function. Prog Lipid Res 2005; 44:303-44. [PMID: 16214218 DOI: 10.1016/j.plipres.2005.08.001] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the last decade or so, it has been realised that membranes do not just have a lipid-bilayer structure in which proteins are embedded or with which they associate. Structures are dynamic and contain areas of heterogeneity which are vital for their formation. In this review, we discuss some of the ways in which these dynamic and heterogeneous structures have implications during stress and in relation to certain human diseases. A particular stress is that of temperature which may instigate adaptation in poikilotherms or appropriate defensive responses during fever in mammals. Recent data emphasise the role of membranes in sensing temperature changes and in controlling a regulatory loop with chaperone proteins. This loop seems to need the existence of specific membrane microdomains and also includes association of chaperone (heat stress) proteins with the membrane. The role of microdomains is then discussed further in relation to various human pathologies such as cardiovascular disease, cancer and neurodegenerative diseases. The concept of modifying membrane lipids (lipid therapy) as a means for treating such pathologies is then introduced. Examples are given when such methods have been shown to have benefit. In order to study membrane microheterogeneity in detail and to elucidate possible molecular mechanisms that account for alteration in membrane function, new methods are needed. In the second part of the review, we discuss ultra-sensitive and ultra-resolution imaging techniques. These include atomic force microscopy, single particle tracking, single particle tracing and various modern fluorescence methods. Finally, we deal with computing simulation of membrane systems. Such methods include coarse-grain techniques and Monte Carlo which offer further advances into molecular dynamics. As computational methods advance they will have more application by revealing the very subtle interactions that take place between the lipid and protein components of membranes - and which are so essential to their function.
Collapse
Affiliation(s)
- Làszló Vigh
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Qi XL, Xiu J, Shan KR, Xiao Y, Gu R, Liu RY, Guan ZZ. Oxidative stress induced by beta-amyloid peptide1–42 is involved in the altered composition of cellular membrane lipids and the decreased expression of nicotinic receptors in human SH-SY5Y neuroblastoma cells. Neurochem Int 2005; 46:613-21. [PMID: 15863239 DOI: 10.1016/j.neuint.2005.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Accepted: 02/18/2005] [Indexed: 10/25/2022]
Abstract
The neurotoxic effects and influence of beta-amyloid peptide (Abeta)(1-42) on membrane lipids and nicotinic acetylcholine receptors (nAChRs) in human SH-SY5Y neuroblastoma cells were investigated in parallel. Exposure of the cultured cells to varying concentrations of Abeta(1-42) evoked a significantly decrease in cellular reduction of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5,diphenyl tetrazolium bromide), together with enhanced lipid peroxidation and protein oxidation. Significant reductions in the total contents of phospholipid and ubiquinone-10, as well as in the levels of the alpha3 and alpha7 subunit proteins of nAChRs were detected in cells exposed to Abeta(1-42). In contrast, such treatment had no effect on the total cellular content of cholesterol. Among these alterations, increased lipid peroxidation and decreased levels of cellular phospholipids were most sensitive to Abeta(1-42), occurring at lower concentrations. In addition, when SH-SY5Y cells were pretreated with the antioxidant Vitamin E, prior to the addition of Abeta(1-42), these alterations in neurotoxicity, oxidative stress, composition of membrane lipids and expression of nAChRs were partially prevented. These findings suggest that stimulation of lipid peroxidation by Abeta may be involved in eliciting the alterations in membrane lipid composition and the reduced expression of nAChRs associated with the pathogenesis of AD.
Collapse
Affiliation(s)
- Xiao-Lan Qi
- Department of Pathology and Molecular Biology, Guiyang Medical College, Guiyang 550004, Guizhou, PR China
| | | | | | | | | | | | | |
Collapse
|