1
|
Wang H, Kc P, Zhang K, Materne C, Lhomme M, Galier S, Ichou F, Neves C, Lehuen A, Haas JT, Salem JE, Guerin M, Lesnik P. MAIT Cells Promote Cholesterol Excretion Pathways Mitigating Atherosclerosis. Circ Res 2025; 136:968-981. [PMID: 40135347 DOI: 10.1161/circresaha.124.325841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous clinical studies have indicated reduced circulating mucosal-associated invariant T (MAIT) cells in individuals with coronary artery disease. However, the precise role and underlying mechanisms of MAIT cells in this context remain unclear. Immune homeostasis plays a pivotal role in the development of atherosclerosis. This study explores the impact of MAIT cells on atherosclerosis. METHODS Vα19+/- Ldlr-/- mice, characterized by a high MAIT cell frequency, and MAIT cell deficient MR1-/- (major histocompatibility complex-related molecule 1) Ldlr-/- mice and their respective controls were used. Starting at 6 weeks of age, mice were subjected to a 1% cholesterol diet for 16 weeks. Additionally, the study analyzed circulating MAIT cell frequency and cholesterol levels in 68 patients with hypercholesterolemia. RESULTS In Vα19+/- Ldlr-/- mice, increased MAIT cells demonstrated a protective effect against atherosclerosis by reducing VLDL-C (very-low-density lipoprotein cholesterol) levels through heightened cholesterol excretion. This effect was accompanied by elevated jejunal ABCB1a, ABCG5, and ABCG8 expression, mediated by augmented levels of Liver X receptor transcription and activation, likely through intestinal IL-22 (interleukin-22) signaling. Conversely, cholesterol reduction mediated by intestinal cholesterol excretion was blocked by inhibition of MAIT cells. Moreover, MAIT cell-deficient MR1-/- Ldlr-/- mice exhibited elevated total cholesterol levels and increased atherosclerotic lesions. In patients with hypercholesterolemia, circulating MAIT cell frequency displayed negative correlations with VLDL-C levels and positive correlations with HDL-C (high-density lipoprotein cholesterol) levels. CONCLUSIONS Our findings demonstrate a new mechanism for plasma VLDL-C clearance by MAIT cell-mediated cholesterol excretion. The results provide further evidence that immunity is involved in cholesterol homeostasis. Targeting intestinal immunity to regulate cholesterol homeostasis holds promise as a new cholesterol-lowering modality to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hua Wang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Pukar Kc
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Kaidi Zhang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Clément Materne
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Sophie Galier
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Carolina Neves
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Agnès Lehuen
- Université Paris Cité, Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR 8104, Inflamex Laboratory, Paris, France (A.L.)
| | - Joel T Haas
- Université de Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, Lille, France (J.T.H.)
| | - Joe-Elie Salem
- INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France (J.-E.S.)
| | - Maryse Guerin
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Philippe Lesnik
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| |
Collapse
|
2
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [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: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
3
|
Loiola RA, Nguyen C, Dib S, Saint-Pol J, Dehouck L, Sevin E, Naudot M, Landry C, Pahnke J, Pot C, Gosselet F. 25-Hydroxycholesterol attenuates tumor necrosis factor alpha-induced blood-brain barrier breakdown in vitro. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167479. [PMID: 39181516 DOI: 10.1016/j.bbadis.2024.167479] [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: 05/24/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Rodrigo Azevedo Loiola
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Cindy Nguyen
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Shiraz Dib
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Julien Saint-Pol
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Lucie Dehouck
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Emmanuel Sevin
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Marie Naudot
- Plateforme d'Ingénierie Cellulaire & Analyses des Protéines ICAP, FR CNRS 3085 ICP, Université de Picardie Jules Verne, F-80039 Amiens, France
| | - Christophe Landry
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO), Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway; Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL), University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany; Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia; School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Ramat Aviv, IL-6997801, Israel
| | - Caroline Pot
- Lausanne University Hospital (CHUV), University of Lausanne, Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, CH-1011 Lausanne, Vaud, Switzerland
| | - Fabien Gosselet
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France.
| |
Collapse
|
4
|
Kumar Nelson V, Jha NK, Nuli MV, Gupta S, Kanna S, Gahtani RM, Hani U, Singh AK, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Al Othaim A, Begum SS, Ahmad F, Mishra PC, Jha SK, Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: Factors and therapeutic implications. Ageing Res Rev 2024; 98:102224. [PMID: 38346505 DOI: 10.1016/j.arr.2024.102224] [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: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that has devastating effects on individuals, often resulting in dementia. AD is primarily defined by the presence of extracellular plaques containing insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein (P-tau). In addition, individuals afflicted by these age-related illnesses experience a diminished state of health, which places significant financial strain on their loved ones. Several risk factors play a significant role in the development of AD. These factors include genetics, diet, smoking, certain diseases (such as cerebrovascular diseases, obesity, hypertension, and dyslipidemia), age, and alcohol consumption. Age-related factors are key contributors to the development of vascular-based neurodegenerative diseases such as AD. In general, the process of aging can lead to changes in the immune system's responses and can also initiate inflammation in the brain. The chronic inflammation and the inflammatory mediators found in the brain play a crucial role in the dysfunction of the blood-brain barrier (BBB). Furthermore, maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. Therefore, in this review, we discussed the role of age and its related factors in the breakdown of the blood-brain barrier and the development of AD. We also discussed the importance of different compounds, such as those with anti-aging properties, and other compounds that can help maintain the integrity of the blood-brain barrier in the prevention of AD. This review builds a strong correlation between age-related factors, degradation of the BBB, and its impact on AD.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sandeep Kanna
- Department of pharmaceutics, Chalapathi Institute of Pharmaceutical Sciences, Chalapathi Nagar, Guntur 522034, India
| | - Reem M Gahtani
- Departement of Clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Arun Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Science, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - S Sabarunisha Begum
- Department of Biotechnology, P.S.R. Engineering College, Sivakasi 626140, India
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh, 13713, Saudi Arabia
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
5
|
Nguyen C, Saint-Pol J, Dib S, Pot C, Gosselet F. 25-Hydroxycholesterol in health and diseases. J Lipid Res 2024; 65:100486. [PMID: 38104944 PMCID: PMC10823077 DOI: 10.1016/j.jlr.2023.100486] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Cholesterol is an essential structural component of all membranes of mammalian cells where it plays a fundamental role not only in cellular architecture, but also, for example, in signaling pathway transduction, endocytosis process, receptor functioning and recycling, or cytoskeleton remodeling. Consequently, intracellular cholesterol concentrations are tightly regulated by complex processes, including cholesterol synthesis, uptake from circulating lipoproteins, lipid transfer to these lipoproteins, esterification, and metabolization into oxysterols that are intermediates for bile acids. Oxysterols have been considered for long time as sterol waste products, but a large body of evidence has clearly demonstrated that they play key roles in central nervous system functioning, immune cell response, cell death, or migration and are involved in age-related diseases, cancers, autoimmunity, or neurological disorders. Among all the existing oxysterols, this review summarizes basic as well as recent knowledge on 25-hydroxycholesterol which is mainly produced during inflammatory or infectious situations and that in turn contributes to immune response, central nervous system disorders, atherosclerosis, macular degeneration, or cancer development. Effects of its metabolite 7α,25-dihydroxycholesterol are also presented and discussed.
Collapse
Affiliation(s)
- Cindy Nguyen
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Julien Saint-Pol
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Shiraz Dib
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Caroline Pot
- Department of Clinical Neurosciences, Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabien Gosselet
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France.
| |
Collapse
|
6
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Dib S, Loiola RA, Sevin E, Saint-Pol J, Shimizu F, Kanda T, Pahnke J, Gosselet F. TNFα Activates the Liver X Receptor Signaling Pathway and Promotes Cholesterol Efflux from Human Brain Pericytes Independently of ABCA1. Int J Mol Sci 2023; 24:ijms24065992. [PMID: 36983062 PMCID: PMC10056409 DOI: 10.3390/ijms24065992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Neuroinflammation and brain lipid imbalances are observed in Alzheimer's disease (AD). Tumor necrosis factor-α (TNFα) and the liver X receptor (LXR) signaling pathways are involved in both processes. However, limited information is currently available regarding their relationships in human brain pericytes (HBP) of the neurovascular unit. In cultivated HBP, TNFα activates the LXR pathway and increases the expression of one of its target genes, the transporter ATP-binding cassette family A member 1 (ABCA1), while ABCG1 is not expressed. Apolipoprotein E (APOE) synthesis and release are diminished. The cholesterol efflux is promoted, but is not inhibited, when ABCA1 or LXR are blocked. Moreover, as for TNFα, direct LXR activation by the agonist (T0901317) increases ABCA1 expression and the associated cholesterol efflux. However, this process is abolished when LXR/ABCA1 are both inhibited. Neither the other ABC transporters nor the SR-BI are involved in this TNFα-mediated lipid efflux regulation. We also report that inflammation increases ABCB1 expression and function. In conclusion, our data suggest that inflammation increases HBP protection against xenobiotics and triggers an LXR/ABCA1 independent cholesterol release. Understanding the molecular mechanisms regulating this efflux at the level of the neurovascular unit remains fundamental to the characterization of links between neuroinflammation, cholesterol and HBP function in neurodegenerative disorders.
Collapse
Affiliation(s)
- Shiraz Dib
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Rodrigo Azevedo Loiola
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Emmanuel Sevin
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Julien Saint-Pol
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube 755-8505, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube 755-8505, Japan
| | - Jens Pahnke
- Department of Pathology, Section of Neuropathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Pahnke Lab (Drug Development and Chemical Biology), Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 3, 1004 Riga, Latvia
- Department of Neurobiology, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| |
Collapse
|
8
|
24-Hydroxycholesterol Induces Tau Proteasome-Dependent Degradation via the SIRT1/PGC1α/Nrf2 Pathway: A Potential Mechanism to Counteract Alzheimer’s Disease. Antioxidants (Basel) 2023; 12:antiox12030631. [PMID: 36978879 PMCID: PMC10044740 DOI: 10.3390/antiox12030631] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Considerable evidence indicates that cholesterol oxidation products, named oxysterols, play a key role in several events involved in Alzheimer’s disease (AD) pathogenesis. Although the majority of oxysterols causes neuron dysfunction and degeneration, 24-hydroxycholesterol (24-OHC) has recently been thought to be neuroprotective also. The present study aimed at supporting this concept by exploring, in SK-N-BE neuroblastoma cells, whether 24-OHC affected the neuroprotective SIRT1/PGC1α/Nrf2 axis. We demonstrated that 24-OHC, through the up-regulation of the deacetylase SIRT1, was able to increase both PGC1α and Nrf2 expression and protein levels, as well as Nrf2 nuclear translocation. By acting on this neuroprotective pathway, 24-OHC favors tau protein clearance by triggering tau ubiquitination and subsequently its degradation through the ubiquitin–proteasome system. We also observed a modulation of SIRT1, PGC1α, and Nrf2 expression and synthesis in the brain of AD patients with the progression of the disease, suggesting their potential role in neuroprotection. These findings suggest that 24-OHC contributes to tau degradation through the up-regulation of the SIRT1/PGC1α/Nrf2 axis. Overall, the evidence points out the importance of avoiding 24-OHC loss, which can occur in the AD brain, and of limiting SIRT1, PGC1α, and Nrf2 deregulation in order to prevent the neurotoxic accumulation of hyperphosphorylated tau and counteract neurodegeneration.
Collapse
|
9
|
Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int J Mol Sci 2023; 24:ijms24032710. [PMID: 36769032 PMCID: PMC9916529 DOI: 10.3390/ijms24032710] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Central nervous system (CNS) disorders represent one of the leading causes of global health burden. Nonetheless, new therapies approved against these disorders are among the lowest compared to their counterparts. The absence of reliable and efficient in vitro blood-brain barrier (BBB) models resembling in vivo barrier properties stands out as a significant roadblock in developing successful therapy for CNS disorders. Therefore, advancement in the creation of robust and sensitive in vitro BBB models for drug screening might allow us to expedite neurological drug development. This review discusses the major in vitro BBB models developed as of now for exploring the barrier properties of the cerebral vasculature. Our main focus is describing existing in vitro models, including the 2D transwell models covering both single-layer and co-culture models, 3D organoid models, and microfluidic models with their construction, permeability measurement, applications, and limitations. Although microfluidic models are better at recapitulating the in vivo properties of BBB than other models, significant gaps still exist for their use in predicting the performance of neurotherapeutics. However, this comprehensive account of in vitro BBB models can be useful for researchers to create improved models in the future.
Collapse
|
10
|
Chai AB, Callaghan R, Gelissen IC. Regulation of P-Glycoprotein in the Brain. Int J Mol Sci 2022; 23:ijms232314667. [PMID: 36498995 PMCID: PMC9740459 DOI: 10.3390/ijms232314667] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Maintenance of the tightly regulated homeostatic environment of the brain is facilitated by the blood-brain barrier (BBB). P-glycoprotein (P-gp), an ATP-binding cassette transporter, is expressed on the luminal surface of the endothelial cells in the BBB, and actively exports a wide variety of substrates to limit exposure of the vulnerable brain environment to waste buildup and neurotoxic compounds. Downregulation of P-gp expression and activity at the BBB have been reported with ageing and in neurodegenerative diseases. Upregulation of P-gp at the BBB contributes to poor therapeutic outcomes due to altered pharmacokinetics of CNS-acting drugs. The regulation of P-gp is highly complex, but unravelling the mechanisms involved may help the development of novel and nuanced strategies to modulate P-gp expression for therapeutic benefit. This review summarises the current understanding of P-gp regulation in the brain, encompassing the transcriptional, post-transcriptional and post-translational mechanisms that have been identified to affect P-gp expression and transport activity.
Collapse
Affiliation(s)
- Amanda B. Chai
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Richard Callaghan
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Ingrid C. Gelissen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: ; Tel.: +61-2-8627-0357
| |
Collapse
|
11
|
Moya ELJ, Lombardo SM, Vandenhaute E, Schneider M, Mysiorek C, Türeli AE, Kanda T, Shimizu F, Sano Y, Maubon N, Gosselet F, Günday-Türeli N, Dehouck MP. Interaction of surfactant coated PLGA nanoparticles with in vitro human brain-like endothelial cells. Int J Pharm 2022; 621:121780. [PMID: 35504427 DOI: 10.1016/j.ijpharm.2022.121780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 11/27/2022]
Abstract
Treatment for CNS related diseases are limited by the difficulty of the drugs to cross the blood-brain barrier (BBB). The functionalization of polymeric nanoparticles (NPs) coated with the surfactants polysorbate 80 (PS80) and poloxamer 188 (P188), have shown promising results as drugs carriers are able to cross the BBB on animal models. In this study, poly(lactide-co-glycolide) (PLGA) NPs coated with PS80 and P188, labelled with a fluorescent dye were tested on human pre-clinical in vitro model to evaluate and compare their uptake profiles, mechanisms of transport and crossing over human brain-like endothelial cells (BLECs) mimicking the human BBB. In addition, these NPs were produced using a method facilitating their reproducible production at high scale, the MicroJet reactor® technology. Results showed that both formulations were biocompatible and able to be internalized within the BLECs in different uptake profiles depending on their coating: P188 NP showed higher internalization capacity than PS80 NP. Both NPs uptakes were ATP-dependent, following more than one endocytosis pathway with colocalization in the early endosomes, ending with a NPs release in the brain compartment. Thus, both surfactant-coated PLGA NPs are interesting formulations for delivery to the brain through the BBB, presenting different uptake profiles.
Collapse
Affiliation(s)
- Elisa L J Moya
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), University of Artois, UR 2465, F-62300 Lens, France
| | - Sonia M Lombardo
- MyBiotech GmbH, Industrie Str. 1B, 66802, Überherrn, Germany; Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany
| | | | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany
| | - Caroline Mysiorek
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), University of Artois, UR 2465, F-62300 Lens, France
| | - Akif E Türeli
- MyBiotech GmbH, Industrie Str. 1B, 66802, Überherrn, Germany
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), University of Artois, UR 2465, F-62300 Lens, France
| | | | - Marie-Pierre Dehouck
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), University of Artois, UR 2465, F-62300 Lens, France.
| |
Collapse
|
12
|
Abstract
ABC transporters are a large family of membrane proteins that transport chemically diverse substrates across the cell membrane. Disruption of transport mechanisms mediated by ABC transporters causes the development of various diseases, including atherosclerosis. Methods: A bioinformatic analysis of a dataset from Gene Expression Omnibus (GEO) was performed. A GEO dataset containing data on gene expression levels in samples of atherosclerotic lesions and control arteries without atherosclerotic lesions from carotid, femoral, and infrapopliteal arteries was used for analysis. To evaluate differentially expressed genes, a bioinformatic analysis was performed in comparison groups using the limma package in R (v. 4.0.2) and the GEO2R and Phantasus tools (v. 1.11.0). Results: The obtained data indicate the differential expression of many ABC transporters belonging to different subfamilies. The differential expressions of ABC transporter genes involved in lipid transport, mechanisms of multidrug resistance, and mechanisms of ion exchange are shown. Differences in the expression of transporters in tissue samples from different arteries are established. Conclusions: The expression of ABC transporter genes demonstrates differences in atherosclerotic samples and normal arteries, which may indicate the involvement of transporters in the pathogenesis of atherosclerosis.
Collapse
|
13
|
Gamba P, Giannelli S, Staurenghi E, Testa G, Sottero B, Biasi F, Poli G, Leonarduzzi G. The Controversial Role of 24-S-Hydroxycholesterol in Alzheimer's Disease. Antioxidants (Basel) 2021; 10:antiox10050740. [PMID: 34067119 PMCID: PMC8151638 DOI: 10.3390/antiox10050740] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 01/19/2023] Open
Abstract
The development of Alzheimer’s disease (AD) is influenced by several events, among which the dysregulation of cholesterol metabolism in the brain plays a major role. Maintenance of brain cholesterol homeostasis is essential for neuronal functioning and brain development. To maintain the steady-state level, excess brain cholesterol is converted into the more hydrophilic metabolite 24-S-hydroxycholesterol (24-OHC), also called cerebrosterol, by the neuron-specific enzyme CYP46A1. A growing bulk of evidence suggests that cholesterol oxidation products, named oxysterols, are the link connecting altered cholesterol metabolism to AD. It has been shown that the levels of some oxysterols, including 27-hydroxycholesterol, 7β-hydroxycholesterol and 7-ketocholesterol, significantly increase in AD brains contributing to disease progression. In contrast, 24-OHC levels decrease, likely due to neuronal loss. Among the different brain oxysterols, 24-OHC is certainly the one whose role is most controversial. It is the dominant oxysterol in the brain and evidence shows that it represents a signaling molecule of great importance for brain function. However, numerous studies highlighted the potential role of 24-OHC in favoring AD development, since it promotes neuroinflammation, amyloid β (Aβ) peptide production, oxidative stress and cell death. In parallel, 24-OHC has been shown to exert several beneficial effects against AD progression, such as preventing tau hyperphosphorylation and Aβ production. In this review we focus on the current knowledge of the controversial role of 24-OHC in AD pathogenesis, reporting a detailed overview of the findings about its levels in different AD biological samples and its noxious or neuroprotective effects in the brain. Given the relevant role of 24-OHC in AD pathophysiology, its targeting could be useful for disease prevention or slowing down its progression.
Collapse
|
14
|
Dib S, Pahnke J, Gosselet F. Role of ABCA7 in Human Health and in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22094603. [PMID: 33925691 PMCID: PMC8124837 DOI: 10.3390/ijms22094603] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Several studies, including genome wide association studies (GWAS), have strongly suggested a central role for the ATP-binding cassette transporter subfamily A member 7 (ABCA7) in Alzheimer’s disease (AD). This ABC transporter is now considered as an important genetic determinant for late onset Alzheimer disease (LOAD) by regulating several molecular processes such as cholesterol metabolism and amyloid processing and clearance. In this review we shed light on these new functions and their cross-talk, explaining its implication in brain functioning, and therefore in AD onset and development.
Collapse
Affiliation(s)
- Shiraz Dib
- UR2465, LBHE-Blood–Brain Barrier Laboratory, University Artois, 62300 Lens, France;
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway;
- LIED, University of Lübeck, Ratzenburger Allee 160, 23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 3, 1004 Riga, Latvia
- Department of Bioorganic Chemistry, Leibniz-Institute of Plant Biochemistry, Weinberg 3, 06120 Halle, Germany
| | - Fabien Gosselet
- UR2465, LBHE-Blood–Brain Barrier Laboratory, University Artois, 62300 Lens, France;
- Correspondence: ; Tel.: +33-(0)3-21791733
| |
Collapse
|
15
|
Hutchinson SA, Websdale A, Cioccoloni G, Røberg-Larsen H, Lianto P, Kim B, Rose A, Soteriou C, Pramanik A, Wastall LM, Williams BJ, Henn MA, Chen JJ, Ma L, Moore JB, Nelson E, Hughes TA, Thorne JL. Liver x receptor alpha drives chemoresistance in response to side-chain hydroxycholesterols in triple negative breast cancer. Oncogene 2021; 40:2872-2883. [PMID: 33742124 PMCID: PMC8062267 DOI: 10.1038/s41388-021-01720-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022]
Abstract
Triple negative breast cancer (TNBC) is challenging to treat successfully because targeted therapies do not exist. Instead, systemic therapy is typically restricted to cytotoxic chemotherapy, which fails more often in patients with elevated circulating cholesterol. Liver x receptors are ligand-dependent transcription factors that are homeostatic regulators of cholesterol, and are linked to regulation of broad-affinity xenobiotic transporter activity in non-tumor tissues. We show that LXR ligands confer chemotherapy resistance in TNBC cell lines and xenografts, and that LXRalpha is necessary and sufficient to mediate this resistance. Furthermore, in TNBC patients who had cancer recurrences, LXRalpha and ligands were independent markers of poor prognosis and correlated with P-glycoprotein expression. However, in patients who survived their disease, LXRalpha signaling and P-glycoprotein were decoupled. These data reveal a novel chemotherapy resistance mechanism in this poor prognosis subtype of breast cancer. We conclude that systemic chemotherapy failure in some TNBC patients is caused by co-opting the LXRalpha:P-glycoprotein axis, a pathway highly targetable by therapies that are already used for prevention and treatment of other diseases.
Collapse
Affiliation(s)
- Samantha A Hutchinson
- School of Food Science and Nutrition, University of Leeds, Leeds, UK.,Institute for Cancer Research, London, UK
| | - Alex Websdale
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | | | | | - Priscilia Lianto
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Baek Kim
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ailsa Rose
- School of Medicine, University of Leeds, Leeds, UK
| | - Chrysa Soteriou
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | | | | | | | - Madeline A Henn
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, Illinois, USA
| | - Joy J Chen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, Illinois, USA
| | - Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, Illinois, USA
| | | | - Erik Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, Illinois, USA.,Cancer Center at Illinois, University of Illinois at Urbana Champaign, Urbana, Illinois, USA.,Division of Nutritional Sciences, University of Illinois at Urbana Champaign, Urbana, Illinois, USA.,University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA.,Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, Illinois, USA
| | - Thomas A Hughes
- School of Medicine, University of Leeds, Leeds, UK. .,Leeds Breast Cancer Research Group, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| | - James L Thorne
- School of Food Science and Nutrition, University of Leeds, Leeds, UK. .,Leeds Breast Cancer Research Group, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| |
Collapse
|
16
|
Gliozzi M, Musolino V, Bosco F, Scicchitano M, Scarano F, Nucera S, Zito MC, Ruga S, Carresi C, Macrì R, Guarnieri L, Maiuolo J, Tavernese A, Coppoletta AR, Nicita C, Mollace R, Palma E, Muscoli C, Belzung C, Mollace V. Cholesterol homeostasis: Researching a dialogue between the brain and peripheral tissues. Pharmacol Res 2020; 163:105215. [PMID: 33007421 DOI: 10.1016/j.phrs.2020.105215] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol homeostasis is a highly regulated process in human body because of its several functions underlying the biology of cell membranes, the synthesis of all steroid hormones and bile acids and the need of trafficking lipids destined to cell metabolism. In particular, it has been recognized that peripheral and central nervous system cholesterol metabolism are separated by the blood brain barrier and are regulated independently; indeed, peripherally, it depends on the balance between dietary intake and hepatic synthesis on one hand and its degradation on the other, whereas in central nervous system it is synthetized de novo to ensure brain physiology. In view of this complex metabolism and its relevant functions in mammalian, impaired levels of cholesterol can induce severe cellular dysfunction leading to metabolic, cardiovascular and neurodegenerative diseases. The aim of this review is to clarify the role of cholesterol homeostasis in health and disease highlighting new intriguing aspects of the cross talk between its central and peripheral metabolism.
Collapse
Affiliation(s)
- Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Saverio Nucera
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Stefano Ruga
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Roberta Macrì
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Annamaria Tavernese
- Division of Cardiology, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Caterina Nicita
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Rocco Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Ernesto Palma
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| | | | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH) - Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy.
| |
Collapse
|
17
|
Xiao M, Xiao ZJ, Yang B, Lan Z, Fang F. Blood-Brain Barrier: More Contributor to Disruption of Central Nervous System Homeostasis Than Victim in Neurological Disorders. Front Neurosci 2020; 14:764. [PMID: 32903669 PMCID: PMC7438939 DOI: 10.3389/fnins.2020.00764] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic but solid shield in the cerebral microvascular system. It plays a pivotal role in maintaining central nervous system (CNS) homeostasis by regulating the exchange of materials between the circulation and the brain and protects the neural tissue from neurotoxic components as well as pathogens. Here, we discuss the development of the BBB in physiological conditions and then focus on the role of the BBB in cerebrovascular disease, including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Finally, we summarize recent advancements in the development of therapies targeting the BBB and outline future directions and outstanding questions in the field. We propose that BBB dysfunction not only results from, but is causal in the pathogenesis of neurological disorders; the BBB is more a contributor to the disruption of CNS homeostasis than a victim in neurological disorders.
Collapse
Affiliation(s)
- Minjia Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Jie Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Yang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ziwei Lan
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Fang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Pardridge WM. The Isolated Brain Microvessel: A Versatile Experimental Model of the Blood-Brain Barrier. Front Physiol 2020; 11:398. [PMID: 32457645 PMCID: PMC7221163 DOI: 10.3389/fphys.2020.00398] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
Abstract
A versatile experimental model for the investigation of the blood-brain barrier (BBB), including the neuro-vascular unit, is the isolated brain microvessel preparation. Brain microvessels are primarily comprised of endothelial cells, but also include pericytes, pre-capillary arteriolar smooth muscle cells, astrocyte foot processes, and occasional nerve endings. These microvessels can be isolated from brain with a 3 h procedure, and the microvessels are free of brain parenchyma. Brain microvessels have been isolated from fresh animal brain, fresh human brain obtained at neurosurgery, as well as fresh or frozen autopsy human brain. Brain microvessels are the starting point for isolation of brain microvessel RNA, which then enables the production of BBB cDNA libraries and a genomics analysis of the brain microvasculature. Brain microvessels, combined with quantitative targeted absolute proteomics, allow for the quantitation of specific transporters or receptors expressed at the brain microvasculature. Brain microvessels, combined with specific antibodies and immune labeling of isolated capillaries, allow for the cellular location of proteins expressed within the neuro-vascular unit. Isolated brain microvessels can be used as an “in vitro” preparation of the BBB for the study of the kinetic parameters of BBB carrier-mediated transport (CMT) systems, or for the determination of dissociation constants of peptide binding to BBB receptor-mediated transport (RMT) systems expressed at either the animal or the human BBB. This review will discuss how the isolated brain microvessel model system has advanced our understanding of the organization and functional properties of the BBB, and highlight recent renewed interest in this 50 year old model of the BBB.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
19
|
Saint-Pol J, Gosselet F, Duban-Deweer S, Pottiez G, Karamanos Y. Targeting and Crossing the Blood-Brain Barrier with Extracellular Vesicles. Cells 2020; 9:cells9040851. [PMID: 32244730 PMCID: PMC7226770 DOI: 10.3390/cells9040851] [Citation(s) in RCA: 312] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022] Open
Abstract
The blood–brain barrier (BBB) is one of the most complex and selective barriers in the human organism. Its role is to protect the brain and preserve the homeostasis of the central nervous system (CNS). The central elements of this physical and physiological barrier are the endothelial cells that form a monolayer of tightly joined cells covering the brain capillaries. However, as endothelial cells regulate nutrient delivery and waste product elimination, they are very sensitive to signals sent by surrounding cells and their environment. Indeed, the neuro-vascular unit (NVU) that corresponds to the assembly of extracellular matrix, pericytes, astrocytes, oligodendrocytes, microglia and neurons have the ability to influence BBB physiology. Extracellular vesicles (EVs) play a central role in terms of communication between cells. The NVU is no exception, as each cell can produce EVs that could help in the communication between cells in short or long distances. Studies have shown that EVs are able to cross the BBB from the brain to the bloodstream as well as from the blood to the CNS. Furthermore, peripheral EVs can interact with the BBB leading to changes in the barrier’s properties. This review focuses on current knowledge and potential applications regarding EVs associated with the BBB.
Collapse
Affiliation(s)
- Julien Saint-Pol
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
- Correspondence: ; Tel.: +33-3-2179-1746
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| | - Sophie Duban-Deweer
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| | - Gwënaël Pottiez
- Caprion Biosciences Inc., 141, Avenue du Président-Kennedy Suite 5650, Montréal, QC H2X3Y7, Canada;
| | - Yannis Karamanos
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, University Artois, F-62300 Lens, France; (F.G.); (S.D.-D.); (Y.K.)
| |
Collapse
|
20
|
Versele R, Corsi M, Fuso A, Sevin E, Businaro R, Gosselet F, Fenart L, Candela P. Ketone Bodies Promote Amyloid-β 1-40 Clearance in a Human in Vitro Blood-Brain Barrier Model. Int J Mol Sci 2020; 21:E934. [PMID: 32023814 PMCID: PMC7037612 DOI: 10.3390/ijms21030934] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the abnormal accumulation of amyloid-β (Aβ) peptides in the brain. The pathological process has not yet been clarified, although dysfunctional transport of Aβ across the blood-brain barrier (BBB) appears to be integral to disease development. At present, no effective therapeutic treatment against AD exists, and the adoption of a ketogenic diet (KD) or ketone body (KB) supplements have been investigated as potential new therapeutic approaches. Despite experimental evidence supporting the hypothesis that KBs reduce the Aβ load in the AD brain, little information is available about the effect of KBs on BBB and their effect on Aβ transport. Therefore, we used a human in vitro BBB model, brain-like endothelial cells (BLECs), to investigate the effect of KBs on the BBB and on Aβ transport. Our results show that KBs do not modify BBB integrity and do not cause toxicity to BLECs. Furthermore, the presence of KBs in the culture media was combined with higher MCT1 and GLUT1 protein levels in BLECs. In addition, KBs significantly enhanced the protein levels of LRP1, P-gp, and PICALM, described to be involved in Aβ clearance. Finally, the combined use of KBs promotes Aβ efflux across the BBB. Inhibition experiments demonstrated the involvement of LRP1 and P-gp in the efflux. This work provides evidence that KBs promote Aβ clearance from the brain to blood in addition to exciting perspectives for studying the use of KBs in therapeutic approaches.
Collapse
Affiliation(s)
- Romain Versele
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Mariangela Corsi
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Dip. di Chirurgia “P. Valdoni”, Via A. Scarpa 16, 00161 Rome, Italy;
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Laurence Fenart
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Pietra Candela
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| |
Collapse
|
21
|
Malaguti M, Cardenia V, Rodriguez-Estrada MT, Hrelia S. Nutraceuticals and physical activity: Their role on oxysterols-mediated neurodegeneration. J Steroid Biochem Mol Biol 2019; 193:105430. [PMID: 31325497 DOI: 10.1016/j.jsbmb.2019.105430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/07/2023]
Abstract
Over the past few years, the contribution of oxysterols to the onset and development of some of the major neurodegenerative diseases (such as Alzheimer's and Parkinson's diseases) has been scientifically asserted, being mainly related to altered brain cholesterol homeostasis. To counteract oxysterol induced inflammation at neuronal level, one possible intervention approach is the administration of some nutrients and/or plant secondary metabolites. On the other hand, the pleiotropic beneficial effects of physical activity seem to play an important role on prevention and counteraction of neurodegenerative diseases, through the modulation of oxysterol homeostasis and the prevention of demyelination. The present review provides a picture of the promising role of nutraceuticals and physical activity on oxysterol-mediated neurodegeneration, pointing out also the different in vitro and in vivo aspects that need to be further investigated for a better understanding of the association of these three counterparts and their overall effect on people at increased risk for neurodegenerative diseases.
Collapse
Affiliation(s)
- Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Rimini, 47921, Italy.
| | - Vladimiro Cardenia
- Department of Agricultural, Forest and Food Sciences DISAFA, University of Turin, Largo Braccini 2, 10095, Grugliasco, Italy
| | | | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Rimini, 47921, Italy
| |
Collapse
|
22
|
Saint-Pol J, Gosselet F. Oxysterols and the NeuroVascular Unit (NVU): A far true love with bright and dark sides. J Steroid Biochem Mol Biol 2019; 191:105368. [PMID: 31026511 DOI: 10.1016/j.jsbmb.2019.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
The brain is isolated from the whole body by the blood-brain barrier (BBB) which is located in brain microvessel endothelial cells (ECs). Through physical and metabolic properties induced by brain pericytes, astrocytes and neurons (these cells and the ECs referred to as the neurovascular unit (NVU)), the BBB hardly restricts exchanges of molecules between the brain and the bloodstream. Among them, cholesterol exchanges between these two compartments are very limited and occur through the transport of LDLs across the BBB. Oxysterols (mainly 24S and 27-hydroxycholesterol) daily cross the BBB and regulate molecule/cholesterol exchanges via Liver X nuclear Receptors (LXRs). In addition, these oxysterols have been linked to pathological processes in neurodegenerative diseases such as Alzheimer's disease. Here we propose an overview of the actual knowledge concerning oxysterols and the NVU cells in physiological and in Alzheimer's disease.
Collapse
Affiliation(s)
- Julien Saint-Pol
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France.
| | - Fabien Gosselet
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France
| |
Collapse
|
23
|
Sato K. [Consideration for future in vitro BBB models - technical development to investigate the drug delivery to the CNS]. Nihon Yakurigaku Zasshi 2019; 152:287-294. [PMID: 30531099 DOI: 10.1254/fpj.152.287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Blood vessels in the central nervous system (CNS) limit the material exchange between blood and parenchyma by blood brain barrier (BBB). At present, no appropriate in vitro BBB models are available for the investigation whether or not the candidate compounds for new drugs could be delivered to the CNS. This causes huge difficulties of the development of CNS drugs and prediction of CNS adverse effects. In this review, I first outline the structures and functions of BBB, together with the parameters used for the quantification of BBB functions. I also introduce the history of in vitro BBB models used in the drug development so far, i.e., the transition from non-cell models to the models using primary culture of rodent cells, porcine, bovine, cell lines, etc. More recently, the application of human cells differentiated from human induced pluripotent stem cells and microfluidic engineering have already started. BBB is essential for the maintenance of brain homeostasis and the mechanisms of the BBB development will be clarified by reproducing functional BBB on the dish. The new in vitro models and the data may provide accurate prediction of drug delivery to the CNS and the improvement of the evaluation system for toxicity and safety, thereby leading to successful launch of new drugs on the market.
Collapse
|
24
|
Testa G, Rossin D, Poli G, Biasi F, Leonarduzzi G. Implication of oxysterols in chronic inflammatory human diseases. Biochimie 2018; 153:220-231. [DOI: 10.1016/j.biochi.2018.06.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/07/2018] [Indexed: 12/18/2022]
|
25
|
Lamartinière Y, Boucau MC, Dehouck L, Krohn M, Pahnke J, Candela P, Gosselet F, Fenart L. ABCA7 Downregulation Modifies Cellular Cholesterol Homeostasis and Decreases Amyloid-β Peptide Efflux in an in vitro Model of the Blood-Brain Barrier. J Alzheimers Dis 2018; 64:1195-1211. [DOI: 10.3233/jad-170883] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Yordenca Lamartinière
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Marie-Christine Boucau
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Lucie Dehouck
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Markus Krohn
- Department of Neuro-/Pathology, University of Oslo (UiO) & Oslo University Hospital (OUS), Oslo, Norway
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo (UiO) & Oslo University Hospital (OUS), Oslo, Norway
- University of Lübeck (UzL), LIED, Lübeck, Germany
- Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
| | - Pietra Candela
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Fabien Gosselet
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Laurence Fenart
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| |
Collapse
|
26
|
Merino-Zamorano C, Fernández-de Retana S, Montañola A, Batlle A, Saint-Pol J, Mysiorek C, Gosselet F, Montaner J, Hernández-Guillamon M. Modulation of Amyloid-β1-40 Transport by ApoA1 and ApoJ Across an in vitro Model of the Blood-Brain Barrier. J Alzheimers Dis 2018; 53:677-91. [PMID: 27232214 DOI: 10.3233/jad-150976] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-β (Aβ) accumulation in Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) is likely caused by the impairment of its brain clearance that partly occurs through the blood-brain barrier (BBB). In this context, an in vitro BBB model is a valuable tool for studying the molecular mechanisms that regulate this process. This study assessed brain Aβ elimination across the BBB and its modulation by the natural chaperones Apolipoprotein A1 (ApoA1) and Apolipoprotein J/Clusterin (ApoJ). The model was based on primary cerebral endothelial cells that were cultured on Matrigel-coated Transwells and treated with fluorescently labeled-Aβ1-40 to track its efflux across the BBB, which corresponds to trafficking from the basolateral (brain) to apical (blood) compartments. We observed that the transport of basolateral Aβ1-40 was enhanced when it was complexed to rApoJ, whereas the complex formed with rApoA1 did not influence Aβ1-40 efflux. However, the presence of rApoA1 in the apical compartment was able to mobilize Aβ1-40 from the basolateral side. We also observed that both rApoA1 and rApoJ moderately crossed the monolayer (from blood to brain) through a mechanism involving the LDL receptor-related protein family. In contrast to the increased rApoJ efflux when complexed to Aβ1-40, rApoA1 trafficking was restricted when it was bound to the Aβ peptide. In summary, the present study highlights the role of ApoJ and ApoA1 in the in vitro modulation of Aβ elimination across the BBB.
Collapse
Affiliation(s)
- Cristina Merino-Zamorano
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sofía Fernández-de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aina Batlle
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Julien Saint-Pol
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Caroline Mysiorek
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Fabien Gosselet
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Neurology, Neurovascular Unit, Vall d'Hebron Hospital, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Coisne C, Tilloy S, Monflier E, Wils D, Fenart L, Gosselet F. Cyclodextrins as Emerging Therapeutic Tools in the Treatment of Cholesterol-Associated Vascular and Neurodegenerative Diseases. Molecules 2016; 21:E1748. [PMID: 27999408 PMCID: PMC6273856 DOI: 10.3390/molecules21121748] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, like atherosclerosis, and neurodegenerative diseases affecting the central nervous system (CNS) are closely linked to alterations of cholesterol metabolism. Therefore, innovative pharmacological approaches aiming at counteracting cholesterol imbalance display promising therapeutic potential. However, these approaches need to take into account the existence of biological barriers such as intestinal and blood-brain barriers which participate in the organ homeostasis and are major defense systems against xenobiotics. Interest in cyclodextrins (CDs) as medicinal agents has increased continuously based on their ability to actively extract lipids from cell membranes and to provide suitable carrier system for drug delivery. Many novel CD derivatives are constantly generated with the objective to improve CD bioavailability, biocompatibility and therapeutic outcomes. Newly designed drug formulation complexes incorporating CDs as drug carriers have demonstrated better efficiency in treating cardiovascular and neurodegenerative diseases. CD-based therapies as cholesterol-sequestrating agent have recently demonstrated promising advances with KLEPTOSE® CRYSMEB in atherosclerosis as well as with the 2-hydroxypropyl-β-cyclodextrin (HPβCD) in clinical trials for Niemann-Pick type C disease. Based on this success, many investigations evaluating the therapeutical beneficial of CDs in Alzheimer's, Parkinson's and Huntington's diseases are currently on-going.
Collapse
Affiliation(s)
- Caroline Coisne
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| | - Sébastien Tilloy
- Unité de Catalyse et de Chimie du Solide (UCCS), University Artois, CNRS, UMR 8181, Lens, F-62300, France.
| | - Eric Monflier
- Unité de Catalyse et de Chimie du Solide (UCCS), University Artois, CNRS, UMR 8181, Lens, F-62300, France.
| | - Daniel Wils
- ROQUETTE, Nutrition & Health R & D, 62136 Lestrem, France.
| | - Laurence Fenart
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| | - Fabien Gosselet
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| |
Collapse
|
28
|
Padala AK, Wani A, Vishwakarma RA, Kumar A, Bharate SB. Functional induction of P-glycoprotein efflux pump by phenyl benzenesulfonamides: Synthesis and biological evaluation of T0901317 analogs. Eur J Med Chem 2016; 122:744-755. [PMID: 27497733 DOI: 10.1016/j.ejmech.2016.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/17/2016] [Accepted: 07/05/2016] [Indexed: 10/21/2022]
Abstract
N-(2,2,2-Trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide (T0901317, 6) is a potent activator of pregnane-X-receptor (PXR), which is a nuclear receptor controlling P-gp expression. Herein, we aimed to investigate P-gp induction activity of T0901317 and establish its structure-activity relationship. T0901317 along with a series of N-triazolyl-methylene-linked benzenesulfonamides were synthesized and screened for P-gp induction activity using a rhodamine-123 based efflux assay in the P-gp overexpressing human adenocarcinoma LS-180 cells, wherein several compounds showed potent P-gp induction activity at 5 μM. Treatment with benzene sulphonamides led to the decrease in intracellular accumulation of a fluorescent P-gp substrate rhodamine-123 up to 48% (control 100%). In the western-blot studies, T0901317 (6) and its triazole linked analog 26e at 5 μM displayed induction of P-gp expression in LS180 cells. These compounds were non-toxic in LS-180 and human neuroblastoma SH-SY5Y cells (IC50 > 50 μM). The compound 26e showed significant P-gp induction even at 0.3 μM, indicating an excellent therapeutic window. These results clearly indicate promise of this class of compounds as potential agents to enhance amyloid-β clearance in Alzheimers patients.
Collapse
Affiliation(s)
- Anil K Padala
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Abubakar Wani
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Ram A Vishwakarma
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Ajay Kumar
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Sandip B Bharate
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| |
Collapse
|
29
|
Kuntz M, Candela P, Saint-Pol J, Lamartinière Y, Boucau MC, Sevin E, Fenart L, Gosselet F. Bexarotene Promotes Cholesterol Efflux and Restricts Apical-to-Basolateral Transport of Amyloid-β Peptides in an In Vitro Model of the Human Blood-Brain Barrier. J Alzheimers Dis 2016; 48:849-62. [PMID: 26402114 DOI: 10.3233/jad-150469] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
One of the prime features of Alzheimer's disease (AD) is the excessive accumulation of amyloid-β (Aβ) peptides in the brain. Several recent studies suggest that this phenomenon results from the dysregulation of cholesterol homeostasis in the brain and impaired bidirectional Aβ exchange between blood and brain. These mechanisms appear to be closely related and are controlled by the blood-brain barrier (BBB) at the brain microvessel level. In animal models of AD, the anticancer drug bexarotene (a retinoid X receptor agonist) has been found to restore cognitive functions and decrease the brain amyloid burden by regulating cholesterol homeostasis. However, the drug's therapeutic effect is subject to debate and the exact mechanism of action has not been characterized. Therefore, the objective of this present study was to determine bexarotene's effects on the BBB. Using an in vitro model of the human BBB, we investigated the drug's effects on cholesterol exchange between abluminal and luminal compartments and the apical-to-basolateral transport of Aβ peptides across the BBB. Our results demonstrated that bexarotene induces the expression of ABCA1 but not ApoE. This upregulation correlates with an increase in ApoE2-, ApoE4-, ApoA-I-, and HDL-mediated cholesterol efflux. Regarding the transport of Aβ peptides, bexarotene increases the expression of ABCB1, which in turn decreases Aβ apical-to-basolateral transport. Our results showed that bexarotene not only promotes the cholesterol exchange between the brain and the blood but also decreases the influx of Aβ peptides across BBB, suggesting that bexarotene is a promising drug candidate for the treatment of AD.
Collapse
|
30
|
Coisne C, Hallier-Vanuxeem D, Boucau MC, Hachani J, Tilloy S, Bricout H, Monflier E, Wils D, Serpelloni M, Parissaux X, Fenart L, Gosselet F. β-Cyclodextrins Decrease Cholesterol Release and ABC-Associated Transporter Expression in Smooth Muscle Cells and Aortic Endothelial Cells. Front Physiol 2016; 7:185. [PMID: 27252658 PMCID: PMC4879322 DOI: 10.3389/fphys.2016.00185] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 05/09/2016] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is an inflammatory disease that leads to an aberrant accumulation of cholesterol in vessel walls forming atherosclerotic plaques. During this process, the mechanism regulating complex cellular cholesterol pools defined as the reverse cholesterol transport (RCT) is altered as well as expression and functionality of transporters involved in this process, namely ABCA1, ABCG1, and SR-BI. Macrophages, arterial endothelial and smooth muscle cells (SMCs) have been involved in the atherosclerotic plaque formation. As macrophages are widely described as the major cell type forming the foam cells by accumulating intracellular cholesterol, RCT alterations have been poorly studied at the arterial endothelial cell and SMC levels. Amongst the therapeutics tested to actively counteract cellular cholesterol accumulation, the methylated β-cyclodextrin, KLEPTOSE® CRYSMEβ, has recently shown promising effects on decreasing the atherosclerotic plaque size in atherosclerotic mouse models. Therefore we investigated in vitro the RCT process occurring in SMCs and in arterial endothelial cells (ABAE) as well as the ability of some modified β-CDs with different methylation degree to modify RCT in these cells. To this aim, cells were incubated in the presence of different methylated β-CDs, including KLEPTOSE® CRYSMEβ. Both cell types were shown to express basal levels of ABCA1 and SR-BI whereas ABCG1 was solely found in ABAE. Upon CD treatments, the percentage of membrane-extracted cholesterol correlated to the methylation degree of the CDs independently of the lipid composition of the cell membranes. Decreasing the cellular cholesterol content with CDs led to reduce the expression levels of ABCA1 and ABCG1. In addition, the cholesterol efflux to ApoA-I and HDL particles was significantly decreased suggesting that cells forming the blood vessel wall are able to counteract the CD-induced loss of cholesterol. Taken together, our observations suggest that methylated β-CDs can significantly reduce the cellular cholesterol content of cells forming atherosclerotic lesions and can subsequently modulate the expression of ABC transporters involved in RCT. The use of methylated β-CDs would represent a valuable and efficient tool to interfere with atherosclerosis pathogenesis in patients, nonetheless their mode of action still needs further investigations to be fully understood and finely controlled at the cellular level.
Collapse
Affiliation(s)
- Caroline Coisne
- EA 2465, Laboratoire de la Barrière Hémato-Encéphalique, Université d'Artois Lens, France
| | | | - Marie-Christine Boucau
- EA 2465, Laboratoire de la Barrière Hémato-Encéphalique, Université d'Artois Lens, France
| | - Johan Hachani
- EA 2465, Laboratoire de la Barrière Hémato-Encéphalique, Université d'Artois Lens, France
| | - Sébastien Tilloy
- Université Artois, CNRS, Centrale Lille, ENSCL, Université Lille, UMR 8181, Unité de Catalyse et de Chimie du Solide (UCCS) Lens, France
| | - Hervé Bricout
- Université Artois, CNRS, Centrale Lille, ENSCL, Université Lille, UMR 8181, Unité de Catalyse et de Chimie du Solide (UCCS) Lens, France
| | - Eric Monflier
- Université Artois, CNRS, Centrale Lille, ENSCL, Université Lille, UMR 8181, Unité de Catalyse et de Chimie du Solide (UCCS) Lens, France
| | - Daniel Wils
- ROQUETTE, Nutrition Direction Lestrem, France
| | | | | | - Laurence Fenart
- EA 2465, Laboratoire de la Barrière Hémato-Encéphalique, Université d'Artois Lens, France
| | - Fabien Gosselet
- EA 2465, Laboratoire de la Barrière Hémato-Encéphalique, Université d'Artois Lens, France
| |
Collapse
|
31
|
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Förster C, Galla HJ, Romero IA, Shusta EV, Stebbins MJ, Vandenhaute E, Weksler B, Brodin B. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab 2016; 36:862-90. [PMID: 26868179 PMCID: PMC4853841 DOI: 10.1177/0271678x16630991] [Citation(s) in RCA: 548] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
The endothelial cells lining the brain capillaries separate the blood from the brain parenchyma. The endothelial monolayer of the brain capillaries serves both as a crucial interface for exchange of nutrients, gases, and metabolites between blood and brain, and as a barrier for neurotoxic components of plasma and xenobiotics. This "blood-brain barrier" function is a major hindrance for drug uptake into the brain parenchyma. Cell culture models, based on either primary cells or immortalized brain endothelial cell lines, have been developed, in order to facilitate in vitro studies of drug transport to the brain and studies of endothelial cell biology and pathophysiology. In this review, we aim to give an overview of established in vitro blood-brain barrier models with a focus on their validation regarding a set of well-established blood-brain barrier characteristics. As an ideal cell culture model of the blood-brain barrier is yet to be developed, we also aim to give an overview of the advantages and drawbacks of the different models described.
Collapse
Affiliation(s)
- Hans C Helms
- Department of Pharmacy, University of Copenhagen, Denmark
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, UK
| | - Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | | | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, HAS, Szeged, Hungary
| | - Carola Förster
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | - Hans J Galla
- Institute of Biochemistry, University of Muenster, Germany
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | | | - Babette Weksler
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, NY, USA
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|
32
|
Gamba P, Testa G, Gargiulo S, Staurenghi E, Poli G, Leonarduzzi G. Oxidized cholesterol as the driving force behind the development of Alzheimer's disease. Front Aging Neurosci 2015; 7:119. [PMID: 26150787 PMCID: PMC4473000 DOI: 10.3389/fnagi.2015.00119] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/03/2015] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD), the most common neurodegenerative disorder associated with dementia, is typified by the pathological accumulation of amyloid Aβ peptides and neurofibrillary tangles (NFT) within the brain. Considerable evidence indicates that many events contribute to AD progression, including oxidative stress, inflammation, and altered cholesterol metabolism. The brain’s high lipid content makes it particularly vulnerable to oxidative species, with the consequent enhancement of lipid peroxidation and cholesterol oxidation, and the subsequent formation of end products, mainly 4-hydroxynonenal and oxysterols, respectively from the two processes. The chronic inflammatory events observed in the AD brain include activation of microglia and astrocytes, together with enhancement of inflammatory molecule and free radical release. Along with glial cells, neurons themselves have been found to contribute to neuroinflammation in the AD brain, by serving as sources of inflammatory mediators. Oxidative stress is intimately associated with neuroinflammation, and a vicious circle has been found to connect oxidative stress and inflammation in AD. Alongside oxidative stress and inflammation, altered cholesterol metabolism and hypercholesterolemia also significantly contribute to neuronal damage and to progression of AD. Increasing evidence is now consolidating the hypothesis that oxidized cholesterol is the driving force behind the development of AD, and that oxysterols are the link connecting the disease to altered cholesterol metabolism in the brain and hypercholesterolemia; this is because of the ability of oxysterols, unlike cholesterol, to cross the blood brain barrier (BBB). The key role of oxysterols in AD pathogenesis has been strongly supported by research pointing to their involvement in modulating neuroinflammation, Aβ accumulation, and cell death. This review highlights the key role played by cholesterol and oxysterols in the brain in AD pathogenesis.
Collapse
Affiliation(s)
- Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| |
Collapse
|
33
|
Pahnke J, Langer O, Krohn M. Alzheimer's and ABC transporters--new opportunities for diagnostics and treatment. Neurobiol Dis 2014; 72 Pt A:54-60. [PMID: 24746857 PMCID: PMC4199932 DOI: 10.1016/j.nbd.2014.04.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 12/26/2022] Open
Abstract
Much has been said about the increasing number of demented patients and the main risk factor 'age'. Frustratingly, we do not know the precise pattern and all modulating factors that provoke the pathologic changes in the brains of affected elderly. We have to diagnose early to be able to stop the progression of diseases that irreversibly destroy brain substance. Familiar AD cases have mislead some researchers for almost 20 years, which has unfortunately narrowed the scientific understanding and has, thus, lead to insufficient funding of independent approaches. Therefore, basic researchers hardly have been able to develop causative treatments and clinicians still do not have access to prognostic and early diagnostic tools. During the recent years it became clear that insufficient Aβ export, physiologically facilitated by the ABC transporter superfamily at the brain's barriers, plays a fundamental role in disease initiation and progression. Furthermore, export mechanisms that are deficient in affected elderly are new targets for activation and, thus, treatment, but ideally also for prevention. In sporadic AD disturbed clearance of β-amyloid from the brain is so far the most important factor for its accumulation in the parenchyma and vessel walls. Here, we review findings about the contribution of ABC transporters and of the perivascular drainage/glymphatic system on β-amyloid clearance. We highlight their potential value for innovative early diagnostics using PET and describe recently described, effective ABC transporter-targeting agents as potential causative treatment for neurodegenerative proteopathies/dementias.
Collapse
Affiliation(s)
- Jens Pahnke
- Neurodegeneration Research Lab (NRL), Department of Neurology, University of Magdeburg, Leipziger Str. 44, Bldg. 64, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44, Bldg. 64, 39120 Magdeburg, Germany.
| | - Oliver Langer
- Health and Environment Department, AIT - Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Währinger-Gürtel 18-20, 1090 Vienna, Austria
| | - Markus Krohn
- Neurodegeneration Research Lab (NRL), Department of Neurology, University of Magdeburg, Leipziger Str. 44, Bldg. 64, 39120 Magdeburg, Germany
| |
Collapse
|
34
|
Campos-Bedolla P, Walter FR, Veszelka S, Deli MA. Role of the Blood–Brain Barrier in the Nutrition of the Central Nervous System. Arch Med Res 2014; 45:610-38. [DOI: 10.1016/j.arcmed.2014.11.018] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
|
35
|
Allinquant B, Clamagirand C, Potier MC. Role of cholesterol metabolism in the pathogenesis of Alzheimer's disease. Curr Opin Clin Nutr Metab Care 2014; 17:319-23. [PMID: 24839952 DOI: 10.1097/mco.0000000000000069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Cholesterol has been shown to stimulate the cleavage of amyloid precursor protein (APP) into amyloid peptides involved in Alzheimer's disease. However, high level of peripheral cholesterol as a risk factor for Alzheimer's disease is still debated. This current review provides an update of the recent literature on cholesterol and APP metabolisms in the brain. RECENT FINDINGS First, a new relationship between neuronal APP and cholesterol has been shown in which this protein controls cholesterol turnover required for neuronal activity. Second, oxysterols are able to stimulate the synthesis of ATP-binding cassette transporters involved in the exchange of amyloid peptides between the blood and the brain. Third, changes in APP targeting to lipid rafts and/or their composition in cholesterol regulate amyloid peptide production. SUMMARY These recent findings open new areas of investigations to control the neuronal activity and to decrease the amyloid peptide levels in brain, opening on new preventive and therapeutic strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Bernadette Allinquant
- aINSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine bInstitut du Cerveau et de la Moelle, UPMC, INSERM UMR S 975, CNRS UMR 7225, Paris, France
| | | | | |
Collapse
|
36
|
Sun JH, Yu JT, Tan L. The Role of Cholesterol Metabolism in Alzheimer’s Disease. Mol Neurobiol 2014; 51:947-65. [DOI: 10.1007/s12035-014-8749-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/07/2014] [Indexed: 12/25/2022]
|
37
|
Testa G, Gamba P, Badilli U, Gargiulo S, Maina M, Guina T, Calfapietra S, Biasi F, Cavalli R, Poli G, Leonarduzzi G. Loading into nanoparticles improves quercetin's efficacy in preventing neuroinflammation induced by oxysterols. PLoS One 2014; 9:e96795. [PMID: 24802026 PMCID: PMC4011877 DOI: 10.1371/journal.pone.0096795] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
Chronic inflammatory events appear to play a fundamental role in Alzheimer's disease (AD)-related neuropathological changes, and to result in neuronal dysfunction and death. The inflammatory responses observed in the AD brain include activation and proliferation of glial cells, together with up-regulation of inflammatory mediators and of free radicals. Along with glial cells, neurons themselves can also react and contribute to neuroinflammatory changes in the AD brain, by serving as sources of inflammatory mediators. Because excess cholesterol cannot be degraded in the brain, it must be excreted from that organ as cholesterol oxidation products (oxysterols), in order to prevent its accumulation. Among risk factors for this neurodegenerative disease, a mechanistic link between altered cholesterol metabolism and AD has been suggested; oxysterols appear to be the missing linkers between the two, because of their neurotoxic effects. This study shows that 24-hydroxycholesterol, 27-hydroxycholesterol, and 7β-hydroxycholesterol, the three oxysterols potentially implicated in AD pathogenesis, induce some pro-inflammatory mediator expression in human neuroblastoma SH-SY5Y cells, via Toll-like receptor-4/cyclooxygenase-2/membrane bound prostaglandin E synthase (TLR4/COX-2/mPGES-1); this clearly indicates that oxysterols may promote neuroinflammatory changes in AD. To confirm this evidence, cells were incubated with the anti-inflammatory flavonoid quercetin; remarkably, its anti-inflammatory effects in SH-SY5Y cells were enhanced when it was loaded into β-cyclodextrin-dodecylcarbonate nanoparticles, versus cells pretreated with free quercetin. The goal of loading quercetin into nanoparticles was to improve its permeation across the blood-brain barrier into the brain, and its bioavailability to reach target cells. The findings show that this drug delivery system might be a new therapeutic strategy for preventing or reducing AD progression.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Ulya Badilli
- Department of Pharmaceutical Technology, University of Ankara, Ankara, Turkey
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Marco Maina
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Tina Guina
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Simone Calfapietra
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
- * E-mail:
| |
Collapse
|
38
|
Gosselet F, Saint-Pol J, Fenart L. Effects of oxysterols on the blood–brain barrier: Implications for Alzheimer’s disease. Biochem Biophys Res Commun 2014; 446:687-91. [DOI: 10.1016/j.bbrc.2013.11.059] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 11/11/2013] [Indexed: 12/31/2022]
|
39
|
Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods Mol Biol 2014; 1175:323-556. [PMID: 25150875 DOI: 10.1007/978-1-4939-0956-8_13] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a major problem of health and disability, with a relevant economic impact on our society. Despite important advances in pathogenesis, diagnosis, and treatment, its primary causes still remain elusive, accurate biomarkers are not well characterized, and the available pharmacological treatments are not cost-effective. As a complex disorder, AD is a polygenic and multifactorial clinical entity in which hundreds of defective genes distributed across the human genome may contribute to its pathogenesis. Diverse environmental factors, cerebrovascular dysfunction, and epigenetic phenomena, together with structural and functional genomic dysfunctions, lead to amyloid deposition, neurofibrillary tangle formation, and premature neuronal death, the major neuropathological hallmarks of AD. Future perspectives for the global management of AD predict that genomics and proteomics may help in the search for reliable biomarkers. In practical terms, the therapeutic response to conventional drugs (cholinesterase inhibitors, multifactorial strategies) is genotype-specific. Genomic factors potentially involved in AD pharmacogenomics include at least five categories of gene clusters: (1) genes associated with disease pathogenesis; (2) genes associated with the mechanism of action of drugs; (3) genes associated with drug metabolism (phase I and II reactions); (4) genes associated with drug transporters; and (5) pleiotropic genes involved in multifaceted cascades and metabolic reactions. The implementation of pharmacogenomic strategies will contribute to optimize drug development and therapeutics in AD and related disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Chair of Genomic Medicine, Camilo José Cela University, 28692, Villanueva de la Cañada, Madrid, Spain,
| | | | | | | | | |
Collapse
|