1
|
Palihati N, Tang Y, Yin Y, Yu D, Liu G, Quan Z, Ni J, Yan Y, Qing H. Clusterin is a Potential Therapeutic Target in Alzheimer's Disease. Mol Neurobiol 2024; 61:3836-3850. [PMID: 38017342 DOI: 10.1007/s12035-023-03801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
In recent years, Clusterin, a glycosylated protein with multiple biological functions, has attracted extensive research attention. It is closely associated with the physiological and pathological states within the organism. Particularly in Alzheimer's disease (AD) research, Clusterin plays a significant role in the disease's occurrence and progression. Numerous studies have demonstrated a close association between Clusterin and AD. Firstly, the expression level of Clusterin in the brain tissue of AD patients is closely related to pathological progression. Secondly, Clusterin is involved in the deposition and formation of β-amyloid, which is a crucial process in AD development. Furthermore, Clusterin may affect the pathogenesis of AD through mechanisms such as regulating inflammation, controlling cell apoptosis, and clearing pathological proteins. Therefore, further research on the relationship between Clusterin and AD will contribute to a deeper understanding of the etiology of this neurodegenerative disease and provide a theoretical basis for developing early diagnostic and therapeutic strategies for AD. This also makes Clusterin one of the research focuses as a potential biomarker for AD diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Nazhakaiti Palihati
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Ding Yu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China.
| |
Collapse
|
2
|
Zhang Y, Luo S, Gao Y, Tong W, Sun S. High-Density Lipoprotein Subfractions Remodeling: A Critical Process for the Treatment of Atherosclerotic Cardiovascular Diseases. Angiology 2024; 75:441-453. [PMID: 36788038 DOI: 10.1177/00033197231157473] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Numerous studies have shown that a low level of high-density lipoprotein cholesterol (HDL-C) is an independent biomarker of cardiovascular disease. High-density lipoprotein (HDL) is considered to be a protective factor for atherosclerosis (AS). Therefore, raising HDL-C has been widely recognized as a promising strategy to treat atherosclerotic cardiovascular diseases (ASCVD). However, several studies have found that increasing HDL-C levels does not necessarily reduce the risk of ASCVD. HDL particles are highly heterogeneous in structure, composition, and biological function. Moreover, HDL particles from atherosclerotic patients exhibit impaired anti-atherogenic functions and these dysfunctional HDL particles might even promote ASCVD. This makes it uncertain that HDL-raising therapy will prevent and treat ASCVD. It is necessary to comprehensively analyze the structure and function of HDL subfractions. We review current advances related to HDL subfractions remodeling and highlight how current lipid-modifying drugs such as niacin, statins, fibrates, and cholesteryl ester transfer protein inhibitors regulate cholesterol concentration of HDL and specific HDL subfractions.
Collapse
Affiliation(s)
- Yaling Zhang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiyu Luo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Gao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, China
| | - Shaowei Sun
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Liu Z, Chao J, Wang C, Sun G, Roeth D, Liu W, Chen X, Li L, Tian E, Feng L, Davtyan H, Blurton-Jones M, Kalkum M, Shi Y. Astrocytic response mediated by the CLU risk allele inhibits OPC proliferation and myelination in a human iPSC model. Cell Rep 2023; 42:112841. [PMID: 37494190 PMCID: PMC10510531 DOI: 10.1016/j.celrep.2023.112841] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/05/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
The C allele of rs11136000 variant in the clusterin (CLU) gene represents the third strongest known genetic risk factor for late-onset Alzheimer's disease. However, whether this single-nucleotide polymorphism (SNP) is functional and what the underlying mechanisms are remain unclear. In this study, the CLU rs11136000 SNP is identified as a functional variant by a small-scale CRISPR-Cas9 screen. Astrocytes derived from isogenic induced pluripotent stem cells (iPSCs) carrying the "C" or "T" allele of the CLU rs11136000 SNP exhibit different CLU expression levels. TAR DNA-binding protein-43 (TDP-43) preferentially binds to the "C" allele to promote CLU expression and exacerbate inflammation. The interferon response and CXCL10 expression are elevated in cytokine-treated C/C astrocytes, leading to inhibition of oligodendrocyte progenitor cell (OPC) proliferation and myelination. Accordingly, elevated CLU and CXCL10 but reduced myelin basic protein (MBP) expression are detected in human brains of C/C carriers. Our study uncovers a mechanism underlying reduced white matter integrity observed in the CLU rs11136000 risk "C" allele carriers.
Collapse
Affiliation(s)
- Zhenqing Liu
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jianfei Chao
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Cheng Wang
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Guihua Sun
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Daniel Roeth
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Wei Liu
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Xianwei Chen
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Li Li
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - E Tian
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lizhao Feng
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Hayk Davtyan
- Department of Neurobiology & Behavior, Institute for Memory Impairments & Neurological Disorders and Sue & Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, Institute for Memory Impairments & Neurological Disorders and Sue & Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
4
|
Gross C, Guérin LP, Socol BG, Germain L, Guérin SL. The Ins and Outs of Clusterin: Its Role in Cancer, Eye Diseases and Wound Healing. Int J Mol Sci 2023; 24:13182. [PMID: 37685987 PMCID: PMC10488069 DOI: 10.3390/ijms241713182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Clusterin (CLU) is a glycoprotein originally discovered in 1983 in ram testis fluid. Rapidly observed in other tissues, it was initially given various names based on its function in different tissues. In 1992, it was finally named CLU by consensus. Nearly omnipresent in human tissues, CLU is strongly expressed at fluid-tissue interfaces, including in the eye and in particular the cornea. Recent research has identified different forms of CLU, with the most prominent being a 75-80 kDa heterodimeric protein that is secreted. Another truncated version of CLU (55 kDa) is localized to the nucleus and exerts pro-apoptotic activities. CLU has been reported to be involved in various physiological processes such as sperm maturation, lipid transportation, complement inhibition and chaperone activity. CLU was also reported to exert important functions in tissue remodeling, cell-cell adhesion, cell-substratum interaction, cytoprotection, apoptotic cell death, cell proliferation and migration. Hence, this protein is sparking interest in tissue wound healing. Moreover, CLU gene expression is finely regulated by cytokines, growth factors and stress-inducing agents, leading to abnormally elevated levels of CLU in many states of cellular disturbance, including cancer and neurodegenerative conditions. In the eye, CLU expression has been reported as being severely increased in several pathologies, such as age-related macular degeneration and Fuch's corneal dystrophy, while it is depleted in others, such as pathologic keratinization. Nevertheless, the precise role of CLU in the development of ocular pathologies has yet to be deciphered. The question of whether CLU expression is influenced by these disorders or contributes to them remains open. In this article, we review the actual knowledge about CLU at both the protein and gene expression level in wound healing, and explore the possibility that CLU is a key factor in cancer and eye diseases. Understanding the expression and regulation of CLU could lead to the development of novel therapeutics for promoting wound healing.
Collapse
Affiliation(s)
- Christelle Gross
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | | | - Bianca G. Socol
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
| | - Lucie Germain
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
5
|
Lobato JCM, Arouche TDS, Nero JD, Filho T, Borges RDS, Neto AMDJC. Interactions between carbon nanotubes and external structures of SARS-CoV-2 using molecular docking and molecular dynamics. J Mol Struct 2023; 1286:135604. [PMID: 37089815 PMCID: PMC10111146 DOI: 10.1016/j.molstruc.2023.135604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Molecular modeling techniques are used to describe the process of interaction between nanotubes and the main structures of the Covid-19 virus: the envelope protein, the main protease, and the Spike glycoprotein. Molecular docking studies show that the ligands have interaction characteristics capable of adsorbing the structures. Molecular dynamics simulations provide information on the mean squared deviation of atomic positions between 0.5 and 3.0 Å. The Gibbs free energy model and solvent accessible surface area approaches are used. Through the results obtained through molecular dynamics simulations, it is noted that the zig-zag nanotube prefers to interact with E-pro, M-pro, and S-gly, respectively. Molecular couplings and free energy showed that the S-gly active site residues strongly interact with zigzag, chiral, and armchair nanotubes, in this order. The interactions demonstrated in this manuscript may predict some promising candidates for virus antagonists, which may be confirmed through experimental approaches.
Collapse
Affiliation(s)
- Júlio Cesar Mendes Lobato
- Laboratory of Preparation and Computation of Nanomaterials (LPCN), Federal University of Pará, C. P. 479, 66075-110, Belém, PA, Brazil
- Proderna, Federal University of Pará, C. P. 479, 66075-110, Belém, PA, Brazil
| | - Tiago da Silva Arouche
- Laboratory of Preparation and Computation of Nanomaterials (LPCN), Federal University of Pará, C. P. 479, 66075-110, Belém, PA, Brazil
| | - Jordan Del Nero
- Physics Faculty, Science Institute of Sciences (ICEN), Federal University of Pará, 66075-110, Belém, PA, Brazil
| | - TarcisoAndrade Filho
- Federal University of the South and Southeast of Pará. 68507-590, Marabá - PA, Brazil
| | - Rosivaldo Dos Santos Borges
- Pharmacy Faculty, Science Institute of Sciences (ICEN), Federal University of Pará, C. P. 479, 66075-110, Belém, PA, Brazil
| | - Antonio Maia de Jesus Chaves Neto
- Laboratory of Preparation and Computation of Nanomaterials (LPCN), Federal University of Pará, C. P. 479, 66075-110, Belém, PA, Brazil
- Physics Faculty, Science Institute of Sciences (ICEN), Federal University of Pará, 66075-110, Belém, PA, Brazil
- Chemistry and Biochemistry, The University of Texas at Arlington, Box 19065, 700 Planetarium Place, Room 130, Arlington, TX 76019-0065
| |
Collapse
|
6
|
Lucas V, Cavadas C, Aveleira CA. Cellular Senescence: From Mechanisms to Current Biomarkers and Senotherapies. Pharmacol Rev 2023; 75:675-713. [PMID: 36732079 DOI: 10.1124/pharmrev.122.000622] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/29/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
An increase in life expectancy in developed countries has led to a surge of chronic aging-related diseases. In the last few decades, several studies have provided evidence of the prominent role of cellular senescence in many of these pathologies. Key traits of senescent cells include cell cycle arrest, apoptosis resistance, and secretome shift to senescence-associated secretory phenotype resulting in increased secretion of various intermediate bioactive factors important for senescence pathophysiology. However, cellular senescence is a highly phenotypically heterogeneous process, hindering the discovery of totally specific and accurate biomarkers. Also, strategies to prevent the pathologic effect of senescent cell accumulation during aging by impairing senescence onset or promoting senescent cell clearance have shown great potential during in vivo studies, and some are already in early stages of clinical translation. The adaptability of these senotherapeutic approaches to human application has been questioned due to the lack of proper senescence targeting and senescence involvement in important physiologic functions. In this review, we explore the heterogeneous phenotype of senescent cells and its influence on the expression of biomarkers currently used for senescence detection. We also discuss the current evidence regarding the efficacy, reliability, development stage, and potential for human applicability of the main existing senotherapeutic strategies. SIGNIFICANCE STATEMENT: This paper is an extensive review of what is currently known about the complex process of cellular senescence and explores its most defining features. The main body of the discussion focuses on how the multifeature fluctuation of the senescence phenotype and the physiological role of cellular senescence have both caused a limitation in the search for truly reliable senescence biomarkers and the progression in the development of senotherapies.
Collapse
Affiliation(s)
- Vasco Lucas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Célia Alexandra Aveleira
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
Trindade D, Cachide M, Soares Martins T, Guedes S, Rosa IM, da Cruz e Silva OA, Henriques AG. Monitoring clusterin and fibrillar structures in aging and dementia. AGING BRAIN 2023; 3:100080. [PMID: 37346145 PMCID: PMC10279921 DOI: 10.1016/j.nbas.2023.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
Objective Clusterin is involved in a variety of physiological processes, including proteostasis. Several clusterin polymorphisms were associated with an increased risk of developing Alzheimer's disease, the world-leading cause of dementia. Herein, the effect of a clusterin polymorphism, aging and dementia in the levels of clusterin in human plasma were analysed in a primary care-based cohort, and the association of this chaperone with fibrillar structures discussed. Methods 64 individuals with dementia (CDR≥1) and 64 age- and sex-matched Controls from a Portuguese cohort were genotyped for CLU rs1136000 polymorphism, and the plasma levels of clusterin and fibrils were assessed. Results An increased prevalence of the CC genotype was observed for the dementia group, although no significant robustness was achieved. CLU rs11136000 SNP did not significantly change plasma clusterin levels in demented individuals. Instead, clusterin levels decreased with aging and even more in individuals with dementia. Importantly, plasma clusterin levels correlated with the presence of fibrillar structures in Control individuals, but not in those with dementia. Conclusion This study reveals a significant decrease in plasma clusterin in demented individuals with aging, which related to altered clusterin-fibrils dynamics. Potentially, plasma clusterin and its association with fibrillar structures can be used to monitor dementia progression along aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ana Gabriela Henriques
- Corresponding author at: Neuroscience and Signaling Group, Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
8
|
Huntoon K, Anderson SK, Ballman KV, Twohy E, Dooley K, Jiang W, An Y, Li J, von Roemeling C, Qie Y, Ross OA, Cerhan JH, Whitton AC, Greenspoon JN, Parney IF, Ashman JB, Bahary JP, Hadjipanayis C, Urbanic JJ, Farace E, Khuntia D, Laack NN, Brown PD, Roberge D, Kim BYS. Association of circulating markers with cognitive decline after radiation therapy for brain metastasis. Neuro Oncol 2023; 25:1123-1131. [PMID: 36472389 PMCID: PMC10237411 DOI: 10.1093/neuonc/noac262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A recent phase III trial (NCT01372774) comparing use of stereotactic radiosurgery [SRS] versus whole-brain radiation therapy [WBRT] after surgical resection of a single brain metastasis revealed that declines in cognitive function were more common with WBRT than with SRS. A secondary endpoint in that trial, and the primary objective in this secondary analysis, was to identify baseline biomarkers associated with cognitive impairment after either form of radiotherapy for brain metastasis. Here we report our findings on APOE genotype and serum levels of associated proteins and their association with radiation-induced neurocognitive decline. METHODS In this retrospective analysis of prospectively collected samples from a completed randomized clinical trial, patients provided blood samples every 3 months that were tested by genotyping and enzyme-linked immunosorbent assay, and results were analyzed in association with cognitive impairment. RESULTS The APOE genotype was not associated with neurocognitive impairment at 3 months. However, low serum levels of ApoJ, ApoE, or ApoA protein (all P < .01) and higher amyloid beta (Aβ 1-42) levels (P = .048) at baseline indicated a greater likelihood of neurocognitive decline at 3 months after SRS, whereas lower ApoJ levels were associated with decline after WBRT (P = .014). CONCLUSIONS Patients with these pretreatment serum markers should be counseled about radiation-related neurocognitive decline.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - S Keith Anderson
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Karla V Ballman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Erin Twohy
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Katharine Dooley
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | - Yi An
- Department of Therapeutic Radiology, Yale-New Haven Hospital, North Haven, Connecticut, USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | | | - Yaqing Qie
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jane H Cerhan
- Department of Psychiatry and Psychology Mayo Clinic, Rochester, Minnesota, USA
| | - Anthony C Whitton
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey N Greenspoon
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathan B Ashman
- Department of Radiation Oncology, Mayo Clinic, Phoenix/Scottsdale, Arizona, USA
| | - Jean-Paul Bahary
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | | | - James J Urbanic
- Department of Radiation Oncology, University of California San Diego, Moores Cancer Center, La Jolla, California, USA
| | - Elana Farace
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Deepak Khuntia
- Department of Radiation Oncology, Precision Cancer Specialists and Varian Medical Systems, Palo Alto, California, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David Roberge
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
9
|
Palma-Lara I, García Alonso-Themann P, Pérez-Durán J, Godínez-Aguilar R, Bonilla-Delgado J, Gómez-Archila D, Espinosa-García AM, Nolasco-Quiroga M, Victoria-Acosta G, López-Ornelas A, Serrano-Bello JC, Olguín-García MG, Palacios-Reyes C. Potential Role of Protein Kinase FAM20C on the Brain in Raine Syndrome, an In Silico Analysis. Int J Mol Sci 2023; 24:ijms24108904. [PMID: 37240249 DOI: 10.3390/ijms24108904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
FAM20C (family with sequence similarity 20, member C) is a serine/threonine-specific protein kinase that is ubiquitously expressed and mainly associated with biomineralization and phosphatemia regulation. It is mostly known due to pathogenic variants causing its deficiency, which results in Raine syndrome (RNS), a sclerosing bone dysplasia with hypophosphatemia. The phenotype is recognized by the skeletal features, which are related to hypophosphorylation of different FAM20C bone-target proteins. However, FAM20C has many targets, including brain proteins and the cerebrospinal fluid phosphoproteome. Individuals with RNS can have developmental delay, intellectual disability, seizures, and structural brain defects, but little is known about FAM20C brain-target-protein dysregulation or about a potential pathogenesis associated with neurologic features. In order to identify the potential FAM20C actions on the brain, an in silico analysis was conducted. Structural and functional defects reported in RNS were described; FAM20C targets and interactors were identified, including their brain expression. Gene ontology of molecular processes, function, and components was completed for these targets, as well as for potential involved signaling pathways and diseases. The BioGRID and Human Protein Atlas databases, the Gorilla tool, and the PANTHER and DisGeNET databases were used. Results show that genes with high expression in the brain are involved in cholesterol and lipoprotein processes, plus axo-dendritic transport and the neuron part. These results could highlight some proteins involved in the neurologic pathogenesis of RNS.
Collapse
Affiliation(s)
- Icela Palma-Lara
- Laboratorio de Morfología Celular y Molecular, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | - Javier Pérez-Durán
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | | | - José Bonilla-Delgado
- Unidad de Investigación, Hospital Regional de Ixtapaluca, Ixtapaluca 56530, Mexico
- Departamento de Biotecnología, Escuela de Ingeniería y Ciencias, Instituto Tecnológico de Monterrey, Toluca de Lerdo 50110, Mexico
| | - Damián Gómez-Archila
- Departamento de Oncología Quirúrgica, Hospital de Gineco-Obstetricia 3, Centro Médico Nacional "La Raza", Ciudad de México 02990, Mexico
| | | | - Manuel Nolasco-Quiroga
- Coordinación de Enseñanza e Investigación, Clínica Hospital Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Huauchinango 73177, Mexico
| | | | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| | - Juan Carlos Serrano-Bello
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | | | - Carmen Palacios-Reyes
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| |
Collapse
|
10
|
Milinkeviciute G, Green KN. Clusterin/apolipoprotein J, its isoforms and Alzheimer's disease. Front Aging Neurosci 2023; 15:1167886. [PMID: 37122381 PMCID: PMC10133478 DOI: 10.3389/fnagi.2023.1167886] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Affiliation(s)
- Giedre Milinkeviciute
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Kim N. Green
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
11
|
Impaired Extracellular Proteostasis in Patients with Heart Failure. Arch Med Res 2023; 54:211-222. [PMID: 36797157 DOI: 10.1016/j.arcmed.2023.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Proteostasis impairment and the consequent increase of amyloid burden in the myocardium have been associated with heart failure (HF) development and poor prognosis. A better knowledge of the protein aggregation process in biofluids could assist the development and monitoring of tailored interventions. AIM To compare the proteostasis status and protein's secondary structures in plasma samples of patients with HF with preserved ejection fraction (HFpEF), patients with HF with reduced ejection fraction (HFrEF), and age-matched individuals. METHODS A total of 42 participants were enrolled in 3 groups: 14 patients with HFpEF, 14 patients with HFrEF, and 14 age-matched individuals. Proteostasis-related markers were analyzed by immunoblotting techniques. Fourier Transform Infrared (FTIR) Spectroscopy in Attenuated Total Reflectance (ATR) was applied to assess changes in the protein's conformational profile. RESULTS Patients with HFrEF showed an elevated concentration of oligomeric proteic species and reduced clusterin levels. ATR-FTIR spectroscopy coupled with multivariate analysis allowed the discrimination of HF patients from age-matched individuals in the protein amide I absorption region (1700-1600 cm-1), reflecting changes in protein conformation, with a sensitivity of 73 and a specificity of 81%. Further analysis of FTIR spectra showed significantly reduced random coils levels in both HF phenotypes. Also, compared to the age-matched group, the levels of structures related to fibril formation were significantly increased in patients with HFrEF, whereas the β-turns were significantly increased in patients with HFpEF. CONCLUSION Both HF phenotypes showed a compromised extracellular proteostasis and different protein conformational changes, suggesting a less efficient protein quality control system.
Collapse
|
12
|
Ko YA, Billheimer JT, Lyssenko NN, Kueider-Paisley A, Wolk DA, Arnold SE, Leung YY, Shaw LM, Trojanowski JQ, Kaddurah-Daouk RF, Kling MA, Rader DJ. ApoJ/Clusterin concentrations are determinants of cerebrospinal fluid cholesterol efflux capacity and reduced levels are associated with Alzheimer's disease. Alzheimers Res Ther 2022; 14:194. [PMID: 36572909 PMCID: PMC9791777 DOI: 10.1186/s13195-022-01119-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/06/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) shares risk factors with cardiovascular disease (CVD) and dysregulated cholesterol metabolism is a mechanism common to both diseases. Cholesterol efflux capacity (CEC) is an ex vivo metric of plasma high-density lipoprotein (HDL) function and inversely predicts incident CVD independently of other risk factors. Cholesterol pools in the central nervous system (CNS) are largely separate from those in blood, and CNS cholesterol excess may promote neurodegeneration. CEC of cerebrospinal fluid (CSF) may be a useful measure of CNS cholesterol trafficking. We hypothesized that subjects with AD and mild cognitive impairment (MCI) would have reduced CSF CEC compared with Cognitively Normal (CN) and that CSF apolipoproteins apoA-I, apoJ, and apoE might have associations with CSF CEC. METHODS We retrieved CSF and same-day ethylenediaminetetraacetic acid (EDTA) plasma from 108 subjects (40 AD; 18 MCI; and 50 CN) from the Center for Neurodegenerative Disease Research biobank at the Perelman School of Medicine, University of Pennsylvania. For CSF CEC assays, we used N9 mouse microglial cells and SH-SY5Y human neuroblastoma cells, and the corresponding plasma assay used J774 cells. Cells were labeled with [3H]-cholesterol for 24 h, had ABCA1 expression upregulated for 6 h, were exposed to 33 μl of CSF, and then were incubated for 2.5 h. CEC was quantified as percent [3H]-cholesterol counts in medium of total counts medium+cells, normalized to a pool sample. ApoA-I, ApoJ, ApoE, and cholesterol were also measured in CSF. RESULTS We found that CSF CEC was significantly lower in MCI compared with controls and was poorly correlated with plasma CEC. CSF levels of ApoJ/Clusterin were also significantly lower in MCI and were significantly associated with CSF CEC. While CSF ApoA-I was also associated with CSF CEC, CSF ApoE had no association with CSF CEC. CSF CEC is significantly and positively associated with CSF Aβ. Taken together, ApoJ/Clusterin may be an important determinant of CSF CEC, which in turn could mitigate risk of MCI and AD risk by promoting cellular efflux of cholesterol or other lipids. In contrast, CSF ApoE does not appear to play a role in determining CSF CEC.
Collapse
Affiliation(s)
- Yi-An Ko
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Jeffrey T. Billheimer
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Nicholas N. Lyssenko
- grid.264727.20000 0001 2248 3398Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Alexandra Kueider-Paisley
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27708 USA
| | - David A. Wolk
- grid.25879.310000 0004 1936 8972Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Steven E. Arnold
- grid.38142.3c000000041936754XDepartment of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Yuk Yee Leung
- grid.25879.310000 0004 1936 8972Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Leslie M. Shaw
- grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - John Q. Trojanowski
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Rima F. Kaddurah-Daouk
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27708 USA ,grid.26009.3d0000 0004 1936 7961Duke Institute for Brain Sciences, Duke University, Durham, NC 27708 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University, Durham, NC 27708 USA
| | - Mitchel A. Kling
- grid.262671.60000 0000 8828 4546Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, 42 E. Laurel Rd., Suite 1800, Stratford, NJ 08084 USA ,grid.25879.310000 0004 1936 8972Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Daniel J. Rader
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| |
Collapse
|
13
|
Holzer M, Ljubojevic-Holzer S, Souza Junior DR, Stadler JT, Rani A, Scharnagl H, Ronsein GE, Marsche G. HDL Isolated by Immunoaffinity, Ultracentrifugation, or Precipitation is Compositionally and Functionally Distinct. J Lipid Res 2022; 63:100307. [PMID: 36511335 PMCID: PMC9720336 DOI: 10.1016/j.jlr.2022.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
The HDL proteome has been widely recognized as an important mediator of HDL function. While a variety of HDL isolation methods exist, their impact on the HDL proteome and its associated function remain largely unknown. Here, we compared three of the most common methods for HDL isolation, namely immunoaffinity (IA), density gradient ultracentrifugation (UC), and dextran-sulfate precipitation (DS), in terms of their effects on the HDL proteome and associated functionalities. We used state-of-the-art mass spectrometry to identify 171 proteins across all three isolation methods. IA-HDL contained higher levels of paraoxonase 1, apoB, clusterin, vitronectin, and fibronectin, while UC-HDL had higher levels of apoA2, apoC3, and α-1-antytrypsin. DS-HDL was enriched with apoA4 and complement proteins, while the apoA2 content was very low. Importantly, size-exclusion chromatography analysis showed that IA-HDL isolates contained subspecies in the size range above 12 nm, which were entirely absent in UC-HDL and DS-HDL isolates. Analysis of these subspecies indicated that they primarily consisted of apoA1, IGκC, apoC1, and clusterin. Functional analysis revealed that paraoxonase 1 activity was almost completely lost in IA-HDL, despite high paraoxonase content. We observed that the elution conditions, using 3M thiocyanate, during IA resulted in an almost complete loss of paraoxonase 1 activity. Notably, the cholesterol efflux capacity of UC-HDL and DS-HDL was significantly higher compared to IA-HDL. Together, our data clearly demonstrate that the isolation procedure has a substantial impact on the composition, subclass distribution, and functionality of HDL. In summary, our data show that the isolation procedure has a significant impact on the composition, subclass distribution and functionality of HDL. Our data can be helpful in the comparison, replication and analysis of proteomic datasets of HDL.
Collapse
Affiliation(s)
- Michael Holzer
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria,BioTechMed Graz, Graz, Austria,For correspondence: Michael Holzer
| | - Senka Ljubojevic-Holzer
- BioTechMed Graz, Graz, Austria,Department of Cardiology, Medical University of Graz, Graz, Austria,Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | | | - Julia T. Stadler
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Alankrita Rani
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Graziella Eliza Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Gunther Marsche
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria,BioTechMed Graz, Graz, Austria
| |
Collapse
|
14
|
The Influence of Clusterin Glycosylation Variability on Selected Pathophysiological Processes in the Human Body. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7657876. [PMID: 36071866 PMCID: PMC9441386 DOI: 10.1155/2022/7657876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022]
Abstract
The present review gathers together the most important information about variability in clusterin molecular structure, its profile, and the degree of glycosylation occurring in human tissues and body fluids in the context of the utility of these characteristics as potential diagnostic biomarkers of selected pathophysiological conditions. The carbohydrate part of clusterin plays a crucial role in many biological processes such as endocytosis and apoptosis. Many pathologies associated with neurodegeneration, carcinogenesis, metabolic diseases, and civilizational diseases (e.g., cardiovascular incidents and male infertility) have been described as causes of homeostasis disturbance, in which the glycan part of clusterin plays a very important role. The results of the discussed studies suggest that glycoproteomic analysis of clusterin may help differentiate the severity of hippocampal atrophy, detect the causes of infertility with an immune background, and monitor the development of cancer. Understanding the mechanism of clusterin (CLU) action and its binding epitopes may enable to indicate new therapeutic goals. The carbohydrate part of clusterin is considered necessary to maintain its proper molecular conformation, structural stability, and proper systemic and/or local biological activity. Taking into account the wide spectrum of CLU action and its participation in many processes in the human body, further studies on clusterin glycosylation variability are needed to better understand the molecular mechanisms of many pathophysiological conditions. They can also provide the opportunity to find new biomarkers and enrich the panel of diagnostic parameters for diseases that still pose a challenge for modern medicine.
Collapse
|
15
|
Wilson MR, Satapathy S, Jeong S, Fini ME. Clusterin, other extracellular chaperones, and eye disease. Prog Retin Eye Res 2022; 89:101032. [PMID: 34896599 PMCID: PMC9184305 DOI: 10.1016/j.preteyeres.2021.101032] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
Proteostasis refers to all the processes that maintain the correct expression level, location, folding and turnover of proteins, essential to organismal survival. Both inside cells and in body fluids, molecular chaperones play key roles in maintaining proteostasis. In this article, we focus on clusterin, the first-recognized extracellular mammalian chaperone, and its role in diseases of the eye. Clusterin binds to and inhibits the aggregation of proteins that are misfolded due to mutations or stresses, clears these aggregating proteins from extracellular spaces, and facilitates their degradation. Clusterin exhibits three main homeostatic activities: proteostasis, cytoprotection, and anti-inflammation. The so-called "protein misfolding diseases" are caused by aggregation of misfolded proteins that accumulate pathologically as deposits in tissues; we discuss several such diseases that occur in the eye. Clusterin is typically found in these deposits, which is interpreted to mean that its capacity as a molecular chaperone to maintain proteostasis is overwhelmed in the disease state. Nevertheless, the role of clusterin in diseases involving such deposits needs to be better defined before therapeutic approaches can be entertained. A more straightforward case can be made for therapeutic use of clusterin based on its proteostatic role as a proteinase inhibitor, as well as its cytoprotective and anti-inflammatory properties. It is likely that clusterin works together in this way with other extracellular chaperones to protect the eye from disease, and we discuss several examples. We end this article by predicting future steps that may lead to development of clusterin as a biological drug.
Collapse
Affiliation(s)
- Mark R Wilson
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Sandeep Satapathy
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Shinwu Jeong
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1333 San Pablo Street., Los Angeles, CA, 90033, USA.
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine; Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences, Tufts University, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
16
|
Abstract
The brain, as one of the most lipid-rich organs, heavily relies on lipid transport and distribution to maintain homeostasis and neuronal function. Lipid transport mediated by lipoprotein particles, which are complex structures composed of apolipoproteins and lipids, has been thoroughly characterized in the periphery. Although lipoproteins in the central nervous system (CNS) were reported over half a century ago, the identification of APOE4 as the strongest genetic risk factor for Alzheimer's disease has accelerated investigation of the biology and pathobiology of lipoproteins in the CNS. This review provides an overview of the different components of lipoprotein particles, in particular apolipoproteins, and their involvements in both physiological functions and pathological mechanisms in the CNS.
Collapse
Affiliation(s)
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| |
Collapse
|
17
|
Pan X. The Roles of Fatty Acids and Apolipoproteins in the Kidneys. Metabolites 2022; 12:metabo12050462. [PMID: 35629966 PMCID: PMC9145954 DOI: 10.3390/metabo12050462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The kidneys are organs that require energy from the metabolism of fatty acids and glucose; several studies have shown that the kidneys are metabolically active tissues with an estimated energy requirement similar to that of the heart. The kidneys may regulate the normal and pathological function of circulating lipids in the body, and their glomerular filtration barrier prevents large molecules or large lipoprotein particles from being filtered into pre-urine. Given the permeable nature of the kidneys, renal lipid metabolism plays an important role in affecting the rest of the body and the kidneys. Lipid metabolism in the kidneys is important because of the exchange of free fatty acids and apolipoproteins from the peripheral circulation. Apolipoproteins have important roles in the transport and metabolism of lipids within the glomeruli and renal tubules. Indeed, evidence indicates that apolipoproteins have multiple functions in regulating lipid import, transport, synthesis, storage, oxidation and export, and they are important for normal physiological function. Apolipoproteins are also risk factors for several renal diseases; for example, apolipoprotein L polymorphisms induce kidney diseases. Furthermore, renal apolipoprotein gene expression is substantially regulated under various physiological and disease conditions. This review is aimed at describing recent clinical and basic studies on the major roles and functions of apolipoproteins in the kidneys.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA;
- Diabetes and Obesity Research Center, NYU Langone Hospital—Long Island, Mineola, New York, NY 11501, USA
| |
Collapse
|
18
|
Yuste-Checa P, Bracher A, Hartl FU. The chaperone Clusterin in neurodegeneration-friend or foe? Bioessays 2022; 44:e2100287. [PMID: 35521968 DOI: 10.1002/bies.202100287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022]
Abstract
Fibrillar protein aggregates are the pathological hallmark of a group of age-dependent neurodegenerative conditions, including Alzheimer's and Parkinson's disease. Aggregates of the microtubule-associated protein Tau are observed in Alzheimer's disease and primary tauopathies. Tau pathology propagates from cell to cell in a prion-like process that is likely subject to modulation by extracellular chaperones such as Clusterin. We recently reported that Clusterin delayed Tau fibril formation but enhanced the activity of Tau oligomers to seed aggregation of endogenous Tau in a cellular model. In contrast, Clusterin inhibited the propagation of α-Synuclein aggregates associated with Parkinson's disease. These findings raise the possibility of a mechanistic link between Clusterin upregulation observed in Alzheimer's disease and the progression of Tau pathology. Here we review the diverse functions of Clusterin in the pathogenesis of neurodegenerative diseases, focusing on evidence that Clusterin may act either as a suppressor or enhancer of pathology.
Collapse
Affiliation(s)
- Patricia Yuste-Checa
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
19
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
20
|
Zheng JJ, Agus JK, Hong BV, Tang X, Rhodes CH, Houts HE, Zhu C, Kang JW, Wong M, Xie Y, Lebrilla CB, Mallick E, Witwer KW, Zivkovic AM. Isolation of HDL by sequential flotation ultracentrifugation followed by size exclusion chromatography reveals size-based enrichment of HDL-associated proteins. Sci Rep 2021; 11:16086. [PMID: 34373542 PMCID: PMC8352908 DOI: 10.1038/s41598-021-95451-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/23/2021] [Indexed: 01/02/2023] Open
Abstract
High-density lipoprotein (HDL) particles have multiple beneficial and cardioprotective roles, yet our understanding of their full structural and functional repertoire is limited due to challenges in separating HDL particles from contaminating plasma proteins and other lipid-carrying particles that overlap HDL in size and/or density. Here we describe a method for isolating HDL particles using a combination of sequential flotation density ultracentrifugation and fast protein liquid chromatography with a size exclusion column. Purity was visualized by polyacrylamide gel electrophoresis and verified by proteomics, while size and structural integrity were confirmed by transmission electron microscopy. This HDL isolation method can be used to isolate a high yield of purified HDL from a low starting plasma volume for functional analyses. This method also enables investigators to select their specific HDL fraction of interest: from the least inclusive but highest purity HDL fraction eluting in the middle of the HDL peak, to pooling all of the fractions to capture the breadth of HDL particles in the original plasma sample. We show that certain proteins such as lecithin cholesterol acyltransferase (LCAT), phospholipid transfer protein (PLTP), and clusterin (CLUS) are enriched in large HDL particles whereas proteins such as alpha-2HS-glycoprotein (A2HSG), alpha-1 antitrypsin (A1AT), and vitamin D binding protein (VDBP) are enriched or found exclusively in small HDL particles.
Collapse
Affiliation(s)
| | - Joanne K Agus
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Brian V Hong
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | | | - Hannah E Houts
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Chenghao Zhu
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Jea Woo Kang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Maurice Wong
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Yixuan Xie
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Emily Mallick
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
21
|
van der Spek SJF, Gonzalez-Lozano MA, Koopmans F, Miedema SSM, Paliukhovich I, Smit AB, Li KW. Age-Dependent Hippocampal Proteomics in the APP/PS1 Alzheimer Mouse Model: A Comparative Analysis with Classical SWATH/DIA and directDIA Approaches. Cells 2021; 10:cells10071588. [PMID: 34202490 PMCID: PMC8304546 DOI: 10.3390/cells10071588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder in the human population, for which there is currently no cure. The cause of AD is unknown; however, the toxic effects of amyloid-β (Aβ) are believed to play a role in its onset. To investigate this, we examined changes in global protein levels in a hippocampal synaptosome fraction of the Amyloid Precursor Protein swe/Presenelin 1 dE9 (APP/PS1) mouse model of AD at 6 and 12 months of age (moa). Data independent acquisition (DIA), or Sequential Window Acquisition of all THeoretical fragment-ion (SWATH), was used for a quantitative label-free proteomics analysis. We first assessed the usefulness of a recently improved directDIA workflow as an alternative to conventional DIA data analysis using a project-specific spectral library. Subsequently, we applied directDIA to the 6- and 12-moa APP/PS1 datasets and applied the Mass Spectrometry Downstream Analysis Pipeline (MS-DAP) for differential expression analysis and candidate discovery. We observed most regulation at 12-moa, in particular of proteins involved in Aβ homeostasis and microglial-dependent processes, like synaptic pruning and the immune response, such as APOE, CLU and C1QA-C. All proteomics data are available via ProteomeXchange with identifier PXD025777.
Collapse
|
22
|
Beyond Lipoprotein(a) plasma measurements: Lipoprotein(a) and inflammation. Pharmacol Res 2021; 169:105689. [PMID: 34033878 PMCID: PMC9247870 DOI: 10.1016/j.phrs.2021.105689] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) [Lp(a)] as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is an apoB100 containing lipoprotein covalently bound to apolipoprotein(a) [apo(a)], a glycoprotein. Plasma Lp(a) levels are to a large extent determined by genetics. Its link to cardiovascular disease (CVD) may be driven by its pro-inflammatory effects, of which its association with oxidized phospholipids (oxPL) bound to Lp(a) is the most studied. Various inflammatory conditions, such as rheumatoid arthritis (RA), systemic lupus erythematosus, acquired immunodeficiency syndrome, and chronic renal failure are associated with high Lp(a) levels. In cases of RA, high Lp(a) levels are reversed by interleukin-6 receptor (IL-6R) blockade by tocilizumab, suggesting a potential role for IL-6 in regulating Lp(a) plasma levels. Elevated levels of IL-6 and IL-6R polymorphisms are associated with CVD. Therapies aimed at lowering apo(a) and thereby reducing plasma Lp(a) levels are in clinical trials. Their results will determine if reductions in apo(a) and Lp(a) decrease cardiovascular outcomes. As we enter this new arena of available treatments, there is a need to improve our understanding of mechanisms. This review will focus on the role of Lp(a) in inflammation and CVD.
Collapse
|
23
|
Bungon T, Haslam C, Damiati S, O'Driscoll B, Whitley T, Davey P, Siligardi G, Charmet J, Awan SA. Graphene FET Sensors for Alzheimer's Disease Protein Biomarker Clusterin Detection. Front Mol Biosci 2021; 8:651232. [PMID: 33869287 PMCID: PMC8044944 DOI: 10.3389/fmolb.2021.651232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
We report on the fabrication and characterisation of graphene field-effect transistor (GFET) biosensors for the detection of Clusterin, a prominent protein biomarker of Alzheimer’s disease (AD). The GFET sensors were fabricated on Si/SiO2 substrate using photolithographic patterning and metal lift-off techniques with evaporated chromium and sputtered gold contacts. Raman Spectroscopy was performed on the devices to determine the quality of the graphene. The GFETs were annealed to improve their performance before the channels were functionalized by immobilising the graphene surface with linker molecules and anti-Clusterin antibodies. Concentration of linker molecules was also independently verified by absorption spectroscopy using the highly collimated micro-beam light of Diamond B23 beamline. The detection was achieved through the binding reaction between the antibody and varying concentrations of Clusterin antigen from 1 to 100 pg/mL, as well as specificity tests using human chorionic gonadotropin (hCG), a glycoprotein risk biomarker of certain cancers. The GFETs were characterized using direct current (DC) 4-probe electrical resistance (4-PER) measurements, which demonstrated a limit of detection of the biosensors to be ∼ 300 fg/mL (4 fM). Comparison with back-gated Dirac voltage shifts with varying concentration of Clusterin show 4-PER measurements to be more accurate, at present, and point to a requirement for further optimisation of the fabrication processes for our next generation of GFET sensors. Thus, we have successfully fabricated a promising set of GFET biosensors for the detection of Clusterin protein biomarker. The developed GFET biosensors are entirely generic and also have the potential to be applied to a variety of other disease detection applications such as Parkinson’s, cancer, and cardiovascular.
Collapse
Affiliation(s)
- Theodore Bungon
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| | - Carrie Haslam
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| | - Samar Damiati
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Benjamin O'Driscoll
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| | - Toby Whitley
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| | - Paul Davey
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| | - Giuliano Siligardi
- Diamond Light Source, Rutherford Appleton Laboratory, Oxfordshire, United Kingdom
| | - Jerome Charmet
- Institute of Digital Healthcare, WMG, University of Warwick, Coventry, United Kingdom
| | - Shakil A Awan
- Wolfson Nanomaterials and Devices Laboratory, School of Engineering, Computing and Mathematics, Faculty of Science and Engineering, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
24
|
Identification of novel substrates of a disintegrin and metalloprotease 17 by specific labeling of surface proteins. Heliyon 2021; 6:e05804. [PMID: 33385093 PMCID: PMC7770542 DOI: 10.1016/j.heliyon.2020.e05804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/05/2020] [Accepted: 12/18/2020] [Indexed: 11/22/2022] Open
Abstract
A disintegrin and metalloprotease 17 (ADAM17) catalyzes the cleavage and release of the ectodomains of its substrates at the cell surface in a process termed ectodomain shedding. However, not all ADAM17 substrates have been identified. Here, we used cell surface protein-specific labeling and proteomic approaches to detect and identify ADAM17 substrates. HeLa cell surface proteins were labeled with a fluorescent dye and cultured with or without TAPI-2, an ADAM17 inhibitor. Labeled proteins released into the culture medium were detected by 2-dimensional gel electrophoresis (2DE). Protein spots showing decreased intensity in response to TAPI-2 were selected as substrates of ADAM17 or their binding proteins, and identified by mass spectrometry. ADAM17 knockdown was preformed to examine the behavior of identified proteins. Of 347 proteins detected by 2DE, 49 showed lower intensity in TAPI-2 (+) than in TAPI-2 (-) samples (p < 0.05), and were considered as candidate substrates of ADAM17. Mass spectrometric analysis of 14 protein spots showing >50% decreased intensity identified clusterin as a novel ADAM17 substrate, in addition to known substrates such as desmoglein-2. Western blot analysis showed that ADAM17 knockdown decreased the levels of clusterin fragments cleaved and released from the cell surface. The results identified clusterin as a novel ADAM17 substrate. The method used to identify clusterin could be used to identify the substrates of other sheddases involved in ectodomain shedding.
Collapse
|
25
|
Marsillach J, Adorni MP, Zimetti F, Papotti B, Zuliani G, Cervellati C. HDL Proteome and Alzheimer's Disease: Evidence of a Link. Antioxidants (Basel) 2020; 9:E1224. [PMID: 33287338 PMCID: PMC7761753 DOI: 10.3390/antiox9121224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Several lines of epidemiological evidence link increased levels of high-density lipoprotein-cholesterol (HDL-C) with lower risk of Alzheimer's disease (AD). This observed relationship might reflect the beneficial effects of HDL on the cardiovascular system, likely due to the implication of vascular dysregulation in AD development. The atheroprotective properties of this lipoprotein are mostly due to its proteome. In particular, apolipoprotein (Apo) A-I, E, and J and the antioxidant accessory protein paraoxonase 1 (PON1), are the main determinants of the biological function of HDL. Intriguingly, these HDL constituent proteins are also present in the brain, either from in situ expression, or derived from the periphery. Growing preclinical evidence suggests that these HDL proteins may prevent the aberrant changes in the brain that characterize AD pathogenesis. In the present review, we summarize and critically examine the current state of knowledge on the role of these atheroprotective HDL-associated proteins in AD pathogenesis and physiopathology.
Collapse
Affiliation(s)
- Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| |
Collapse
|
26
|
Bradley D. Clusterin as a Potential Biomarker of Obesity-Related Alzheimer's Disease Risk. Biomark Insights 2020; 15:1177271920964108. [PMID: 33110346 PMCID: PMC7555556 DOI: 10.1177/1177271920964108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/03/2023] Open
Abstract
Over 35% of the adult US population is obese. In turn, excess adiposity increases the risk of multiple complications including type 2 diabetes (T2D), insulin resistance, and cardiovascular disease; yet, obesity also independently heightens risk of Alzheimer's Disease (AD), even after adjusting for other important confounding risk factors including blood pressure, sociodemographics, cholesterol levels, smoking status, and Apolipoprotein E (ApoE) genotype. Among patients over the age of 65 with dementia, 37% have coexisting diabetes, and an estimated 7.3% of cases of AD are directly attributable to midlife obesity. Clusterin, also known as apolipoprotein J (ApoJ), is a multifunctional glycoprotein that acts as a molecular chaperone, assisting folding of secreted proteins. Clusterin has been implicated in several physiological and pathological states, including AD, metabolic disease, and cardiovascular disease. Despite long-standing interest in elucidating clusterin's relationship with amyloid beta (Aβ) aggregation/clearance and toxicity, significant knowledge gaps still exist. Altered clusterin expression and protein levels have been linked with cognitive and memory function, disrupted central nervous system lipid flux, as well as pathogenic brain structure; and its role in cardiometabolic disease suggests that it may be a link between insulin resistance, dyslipidemia, and AD. Here, we briefly highlight clusterin's relevance to AD by presenting existing evidence linking clusterin to AD and cardiometabolic disease, and discussing its potential utility as a biomarker for AD in the presence of obesity-related metabolic disease.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
27
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
28
|
Glomerular clusterin expression is increased in diabetic nephropathy and protects against oxidative stress-induced apoptosis in podocytes. Sci Rep 2020; 10:14888. [PMID: 32913257 PMCID: PMC7484791 DOI: 10.1038/s41598-020-71629-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
Clusterin, a glycoprotein encoded by the CLU gene, is expressed in many tissues, including the kidney, and clusterin expression is upregulated in the glomeruli of patients with various forms of kidney disease. Here, we investigated the role of clusterin in diabetic nephropathy (DN). In this study, we found that glomerular clusterin expression was increased in both patients with DN and streptozotocin-induced diabetic mice and that it co-localised with the podocyte marker WT1, indicating clusterin is expressed in podocytes. In our in vitro analysis, we found no significant change in CLU mRNA expression in podocytes following stimulation with high glucose and angiotensin II; in contrast, CLU mRNA expression was significantly upregulated following methylglyoxal stimulation. Methylglyoxal treatment also significantly decreased the mRNA expression of the slit diaphragm markers ZO-1 and NEPH1 and significantly increased the mRNA expression of the oxidative stress marker HO-1. Lastly, we showed that pre-incubating podocytes with recombinant human clusterin protein increased podocyte survival, prevented slit diaphragm damage, and reduced oxidative stress‒induced apoptosis following methylglyoxal stimulation. Taken together, our results indicate that glomerular clusterin is upregulated in DN, and this increase in clusterin expression may protect against oxidative stress-induced apoptosis in podocytes, providing a possible new therapeutic target for DN and other kidney diseases.
Collapse
|
29
|
Ha J, Moon MK, Kim H, Park M, Cho SY, Lee J, Lee JY, Kim E. Plasma Clusterin as a Potential Link Between Diabetes and Alzheimer Disease. J Clin Endocrinol Metab 2020; 105:5860166. [PMID: 32561922 DOI: 10.1210/clinem/dgaa378] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/13/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Plasma clusterin, a promising biomarker of Alzheimer disease (AD), has been associated with diabetes mellitus (DM). However, clusterin has not been investigated considering a relationship with both DM and AD. In this study, we aimed to investigate the individual and interactive relationships of plasma clusterin levels with both diseases. DESIGN Cross-sectional observation study. METHODS We classified participants by the severity of cognitive (normal cognition, mild cognitive impairment [MCI], and AD) and metabolic (healthy control, prediabetes, and DM) impairments. We evaluated the cognitive and metabolic functions of the participants with neuropsychological assessments, brain magnetic resonance imaging, and various blood tests, to explore potential relationships with clusterin. RESULTS Plasma clusterin levels were higher in participants with AD and metabolic impairment (prediabetes and DM). A two-way ANCOVA revealed no synergistic, but an additive effect of AD and DM on clusterin. Clusterin was negatively correlated with cognitive scores. It was also associated with metabolic status indicated by glycated hemoglobin A1c (HbA1c), the Homeostatic Model Assessment for Insulin Resistance index, and fasting C-peptide. It showed correlations between medial temporal atrophy and periventricular white matter lesions, indicating neurodegeneration and microvascular insufficiency, respectively. Further mediation analysis to understand the triadic relationship between clusterin, AD, and DM revealed that the association between DM and AD was significant when clusterin is considered as a mediator of their relationship. CONCLUSIONS Clusterin is a promising biomarker of DM as well as of AD. Additionally, our data suggest that clusterin may have a role in linking DM with AD as a potential mediator.
Collapse
Affiliation(s)
- Junghee Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Kyong Moon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsun Park
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jimin Lee
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry and Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eosu Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
30
|
Lee J, Hong SW, Kwon H, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Resveratrol, an activator of SIRT1, improves ER stress by increasing clusterin expression in HepG2 cells. Cell Stress Chaperones 2019; 24:825-833. [PMID: 31183612 PMCID: PMC6629741 DOI: 10.1007/s12192-019-01012-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022] Open
Abstract
Endoplasmic reticulum stress (ER stress) is involved in lipid metabolism and lipotoxicity and can lead to apoptosis. Resveratrol, a sirtuin 1 (SIRT1) agonist, prevents ER stress and improves ER stress-induced hepatic steatosis and cell death. Clusterin is a secreted chaperone and has roles in various physiological processes. However, changes in the expression of clusterin upon ER stress and the connection between SIRT1 and clusterin in protection against ER stress are not well known. In cells treated with tunicamycin, resveratrol increased the expression of clusterin mRNA and protein and the secreted clusterin protein level in conditioned medium. Resveratrol decreased protein expression of the ER stress markers, p-PERK, p-IRE1α, and CHOP, and increased the expression of the ER-associated degradation (ERAD) factors, SEL1L and HRD1, in tunicamycin-treated cells. However, no changes in the expression of these genes were observed in clusterin siRNA-transfected cells. Moreover, increased LAMP2 and LC3 expression and decreased Rubicon expression were observed in cells treated with resveratrol or secreted clusterin. These data suggest that SIRT1 activation by resveratrol attenuates ER stress by promoting protective processes such as ERAD and autophagy pathways and that these protective effects are mediated by clusterin.
Collapse
Affiliation(s)
- Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Eun-Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Cheol-Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Ki-Won Oh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Sung-Woo Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Won-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| |
Collapse
|
31
|
de Retana SF, Marazuela P, Solé M, Colell G, Bonaterra A, Sánchez-Quesada JL, Montaner J, Maspoch D, Cano-Sarabia M, Hernández-Guillamon M. Peripheral administration of human recombinant ApoJ/clusterin modulates brain beta-amyloid levels in APP23 mice. ALZHEIMERS RESEARCH & THERAPY 2019; 11:42. [PMID: 31077261 PMCID: PMC6511153 DOI: 10.1186/s13195-019-0498-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Background ApoJ/clusterin is a multifunctional protein highly expressed in the brain. The implication of ApoJ in β-amyloid (Aβ) fibrillization and clearance in the context of Alzheimer’s disease has been widely studied, although the source and concentration of ApoJ that promotes or inhibits Aβ cerebral accumulation is not clear yet. ApoJ is abundant in plasma and approximately 20% can appear bound to HDL-particles. In this regard, the impact of plasmatic ApoJ and its lipidation status on cerebral β-amyloidosis is still not known. Hence, our main objective was to study the effect of a peripheral increase of free ApoJ or reconstituted HDL particles containing ApoJ in an experimental model of cerebral β-amyloidosis. Methods Fourteen-month-old APP23 transgenic mice were subjected to subchronic intravenous treatment with rHDL-rApoJ nanodiscs or free rApoJ for 1 month. Aβ concentration and distribution in the brain, as well as Aβ levels in plasma and CSF, were determined after treatments. Other features associated to AD pathology, such as neuronal loss and neuroinflammation, were also evaluated. Results Both ApoJ-based treatments prevented the Aβ accumulation in cerebral arteries and induced a decrease in total brain insoluble Aβ42 levels. The peripheral treatment with rApoJ also induced an increase in the Aβ40 levels in CSF, whereas the concentration remained unaltered in plasma. At all the endpoints studied, the lipidation of rApoJ did not enhance the protective properties of free rApoJ. The effects obtained after subchronic treatment with free rApoJ were accompanied by a reduction in hippocampal neuronal loss and an enhancement of the expression of a phagocytic marker in microglial cells surrounding Aβ deposits. Finally, despite the activation of this phagocytic phenotype, treatments did not induce a global neuroinflammatory status. In fact, free rApoJ treatment was able to reduce the levels of interleukin-17 (IL17) and keratinocyte chemoattractant (KC) chemokine in the brain. Conclusions Our results demonstrate that an increase in circulating human rApoJ induces a reduction of insoluble Aβ and CAA load in the brain of APP23 mice. Thus, our study suggests that peripheral interventions, based on treatments with multifunctional physiological chaperones, offer therapeutic opportunities to regulate the cerebral Aβ load. Electronic supplementary material The online version of this article (10.1186/s13195-019-0498-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofía Fernández de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Guillem Colell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Anna Bonaterra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBER of Diabetes and Metabolism (CIBERDEM), ISCIII, Madrid, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain.
| |
Collapse
|
32
|
Clusterin silencing restores myoblasts viability and down modulates the inflammatory process in osteoporotic disease. J Transl Med 2019; 17:118. [PMID: 30967152 PMCID: PMC6457035 DOI: 10.1186/s12967-019-1868-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/30/2019] [Indexed: 12/13/2022] Open
Abstract
Background Targeting new molecular pathways leading to Osteoporosis (OP) and Osteoarthritis (OA) is a hot topic for drug discovery. Clusterin (CLU) is a glycoprotein involved in inflammation, proliferation, cell death, neoplastic disease, Alzheimer disease and aging. The present study focuses on the expression and the role of CLU in influencing the decrease of muscle mass and fiber senescence in OP-OA condition. Methods Vastus lateralis muscle biopsies were collected from 20 women with OP undergoing surgery for fragility hip fracture and 20 women undergoing arthroplasty for hip osteoarthritis. Results We found an overexpression of CLU in degenerated fibers in OP closely correlated with interleukin 6 (IL6) and histone H4 acetylation level. Conversely, in OA muscle tissues we observed a weak expression of CLU but no nuclear histone H4 acetylation. Ex vivo studies on isolated human myoblasts confirmed CLU overexpression in OP as compared to OA (p < 0.001). CLU treatment of isolated OP and OA myoblasts showed: modulation of proliferation, morphological changes, increase of histone H4 acetylation and induction of myogenin (MYOG) activation in OP myoblast only. In OP condition, functional knockdown of CLU by siRNA restores proliferative myoblasts capability and tissue damage repair, carried out by an evident upregulation of Transglutaminase 2 (TGM2). We also observed downmodulation of CX3CR1 expression with consequent impairing of the inflammatory infiltrate recruitment. Conclusions Results obtained suggest a potential role of CLU in OP by influencing myoblasts terminal differentiation, epigenetic regulation of muscle cell differentiation and senescence. Moreover, CLU silencing points out its role in the modulation of tissue damage repair and inflammation, proposing it as a new diagnostic marker for muscle degeneration and a potential target for specific therapeutic intervention in OP related sarcopenia.
Collapse
|
33
|
Foster EM, Dangla-Valls A, Lovestone S, Ribe EM, Buckley NJ. Clusterin in Alzheimer's Disease: Mechanisms, Genetics, and Lessons From Other Pathologies. Front Neurosci 2019; 13:164. [PMID: 30872998 PMCID: PMC6403191 DOI: 10.3389/fnins.2019.00164] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Clusterin (CLU) or APOJ is a multifunctional glycoprotein that has been implicated in several physiological and pathological states, including Alzheimer's disease (AD). With a prominent extracellular chaperone function, additional roles have been discussed for clusterin, including lipid transport and immune modulation, and it is involved in pathways common to several diseases such as cell death and survival, oxidative stress, and proteotoxic stress. Although clusterin is normally a secreted protein, it has also been found intracellularly under certain stress conditions. Multiple hypotheses have been proposed regarding the origin of intracellular clusterin, including specific biogenic processes leading to alternative transcripts and protein isoforms, but these lines of research are incomplete and contradictory. Current consensus is that intracellular clusterin is most likely to have exited the secretory pathway at some point or to have re-entered the cell after secretion. Clusterin's relationship with amyloid beta (Aβ) has been of great interest to the AD field, including clusterin's apparent role in altering Aβ aggregation and/or clearance. Additionally, clusterin has been more recently identified as a mediator of Aβ toxicity, as evidenced by the neuroprotective effect of CLU knockdown and knockout in rodent and human iPSC-derived neurons. CLU is also the third most significant genetic risk factor for late onset AD and several variants have been identified in CLU. Although the exact contribution of these variants to altered AD risk is unclear, some have been linked to altered CLU expression at both mRNA and protein levels, altered cognitive and memory function, and altered brain structure. The apparent complexity of clusterin's biogenesis, the lack of clarity over the origin of the intracellular clusterin species, and the number of pathophysiological functions attributed to clusterin have all contributed to the challenge of understanding the role of clusterin in AD pathophysiology. Here, we highlight clusterin's relevance to AD by discussing the evidence linking clusterin to AD, as well as drawing parallels on how the role of clusterin in other diseases and pathways may help us understand its biological function(s) in association with AD.
Collapse
Affiliation(s)
| | | | | | | | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Horodyska J, Reyer H, Wimmers K, Trakooljul N, Lawlor PG, Hamill RM. Transcriptome analysis of adipose tissue from pigs divergent in feed efficiency reveals alteration in gene networks related to adipose growth, lipid metabolism, extracellular matrix, and immune response. Mol Genet Genomics 2018; 294:395-408. [PMID: 30483895 DOI: 10.1007/s00438-018-1515-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 11/13/2018] [Indexed: 12/14/2022]
Abstract
Adipose tissue is hypothesized to play a vital role in regulation of feed efficiency (FE; efficiency in converting energy and nutrients into tissue), of which improvement will simultaneously reduce environmental impact and feed cost per pig. The objective of the present study was to sequence the subcutaneous adipose tissue transcriptome in FE-divergent pigs (n = 16) and identify relevant biological processes underpinning observed differences in FE. We previously demonstrated that high-FE pigs were associated with lower fatness when compared to their counterparts. Here, ontology analysis of a total of 209 annotated genes that were differentially expressed at a p < 0.01 revealed establishment of a dense extracellular matrix and inhibition of capillary formation as one underlying mechanism to achieve suppressed adipogenesis. Moreover, mechanisms ensuring an efficient utilization of lipids in high-FE pigs might be orchestrated by upstream regulators including CEBPA and EGF. Consequently, high-FE adipose tissue could exhibit more efficient cholesterol disposal, whilst inhibition of inflammatory and immune response in high-FE pigs may be an indicator of an optimally functioning adipose tissue. Taken together, adipose tissue growth, extracellular matrix formation, lipid metabolism and inflammatory and immune response are key biological events underpinning the differences in FE. Further investigations focusing on elucidating these processes would assist the animal production industry in optimizing strategies related to nutrient utilization and product quality.
Collapse
Affiliation(s)
- Justyna Horodyska
- Teagasc, Food Research Centre, Ashtown, Dublin 15, Ireland.,Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Henry Reyer
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany.,Faculty of Agricultural and Environmental Sciences, University Rostock, Rostock, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Peadar G Lawlor
- Teagasc, Pig Development Department, AGRIC, Moorepark, Fermoy, Co. Cork, Ireland
| | - Ruth M Hamill
- Teagasc, Food Research Centre, Ashtown, Dublin 15, Ireland.
| |
Collapse
|
35
|
Dlugosz P, Nimpf J. The Reelin Receptors Apolipoprotein E receptor 2 (ApoER2) and VLDL Receptor. Int J Mol Sci 2018; 19:E3090. [PMID: 30304853 PMCID: PMC6213145 DOI: 10.3390/ijms19103090] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 01/28/2023] Open
Abstract
Apolipoprotein E receptor 2 (ApoER2) and VLDL receptor belong to the low density lipoprotein receptor family and bind apolipoprotein E. These receptors interact with the clathrin machinery to mediate endocytosis of macromolecules but also interact with other adapter proteins to perform as signal transduction receptors. The best characterized signaling pathway in which ApoER2 and VLDL receptor (VLDLR) are involved is the Reelin pathway. This pathway plays a pivotal role in the development of laminated structures of the brain and in synaptic plasticity of the adult brain. Since Reelin and apolipoprotein E, are ligands of ApoER2 and VLDLR, these receptors are of interest with respect to Alzheimer's disease. We will focus this review on the complex structure of ApoER2 and VLDLR and a recently characterized ligand, namely clusterin.
Collapse
Affiliation(s)
- Paula Dlugosz
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University Vienna, 1030 Vienna, Austria.
| | - Johannes Nimpf
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University Vienna, 1030 Vienna, Austria.
| |
Collapse
|
36
|
Choi JH, Jeong E, Youn BS, Kim MS. Distinct Ultradian Rhythms in Plasma Clusterin Concentrations in Lean and Obese Korean Subjects. Endocrinol Metab (Seoul) 2018; 33:245-251. [PMID: 29766682 PMCID: PMC6021305 DOI: 10.3803/enm.2018.33.2.245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Blood levels of many hormones show rhythmic fluctuations with variable duration of cycles. Clusterin/apolipoprotein J is a glycoprotein which is highly expressed in the plasma and has modulatory roles in immune and inflammatory reactions, neurobiology, lipid metabolism, and leptin signaling. In this study, we examined the diurnal fluctuations of plasma clusterin concentrations in lean and obese young men. METHODS For the study, 14 subjects (five lean and five obese men; two lean and two obese women) were admitted to the research ward and blood samples were drawn every 30 minutes during light-on period (6:00 AM to 10:00 PM) and every hour during light-off period. RESULTS Notably, plasma clusterin concentrations displayed a unique ultradian rhythm with five cycles a day in both men and women. During the light-on period, circulating clusterin levels showed fluctuating curves with 4 hours regular intervals with sharp peaks and troughs. In contrast, single oscillation curve during light-off exhibited a smoothened/lower peak and longer (8-hour) duration. In obese men, these cycles were phase-advanced by approximately 1 hour, and had reduced amplitude of fluctuating curves and blunted diurnal pattern. Cyclic fluctuations of plasma clusterin were preserved under fasting and unexpected meal condition, suggesting that rhythmic oscillations in plasma clusterin levels are not generated by meal-related cues. CONCLUSION These findings firstly demonstrate a novel pattern of plasma clusterin fluctuations with extremely regular cycles.
Collapse
Affiliation(s)
- Jong Han Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eunheui Jeong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | | | - Min Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
37
|
Gulati T, Huang C, Caramia F, Raghu D, Paul PJ, Goode RJA, Keam SP, Williams SG, Haupt S, Kleifeld O, Schittenhelm RB, Gamell C, Haupt Y. Proteotranscriptomic Measurements of E6-Associated Protein (E6AP) Targets in DU145 Prostate Cancer Cells. Mol Cell Proteomics 2018; 17:1170-1183. [PMID: 29463595 DOI: 10.1074/mcp.ra117.000504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/18/2018] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer is a common cause of cancer-related death in men. E6AP (E6-Associated Protein), an E3 ubiquitin ligase and a transcription cofactor, is elevated in a subset of prostate cancer patients. Genetic manipulations of E6AP in prostate cancer cells expose a role of E6AP in promoting growth and survival of prostate cancer cells in vitro and in vivo However, the effect of E6AP on prostate cancer cells is broad and it cannot be explained fully by previously identified tumor suppressor targets of E6AP, promyelocytic leukemia protein and p27. To explore additional players that are regulated downstream of E6AP, we combined a transcriptomic and proteomic approach. We identified and quantified 16,130 transcripts and 7,209 proteins in castration resistant prostate cancer cell line, DU145. A total of 2,763 transcripts and 308 proteins were significantly altered on knockdown of E6AP. Pathway analyses supported the known phenotypic effects of E6AP knockdown in prostate cancer cells and in parallel exposed novel potential links of E6AP with cancer metabolism, DNA damage repair and immune response. Changes in expression of the top candidates were confirmed using real-time polymerase chain reaction. Of these, clusterin, a stress-induced chaperone protein, commonly deregulated in prostate cancer, was pursued further. Knockdown of E6AP resulted in increased clusterin transcript and protein levels in vitro and in vivo Concomitant knockdown of E6AP and clusterin supported the contribution of clusterin to the phenotype induced by E6AP. Overall, results from this study provide insight into the potential biological pathways controlled by E6AP in prostate cancer cells and identifies clusterin as a novel target of E6AP.
Collapse
Affiliation(s)
- Twishi Gulati
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Cheng Huang
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Franco Caramia
- §Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dinesh Raghu
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Piotr J Paul
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Robert J A Goode
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Simon P Keam
- §Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Scott G Williams
- ‖Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sue Haupt
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Oded Kleifeld
- **Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ralf B Schittenhelm
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Cristina Gamell
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ygal Haupt
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; .,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,‡‡Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,§§Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia.,¶¶Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Loss of clusterin shifts amyloid deposition to the cerebrovasculature via disruption of perivascular drainage pathways. Proc Natl Acad Sci U S A 2017; 114:E6962-E6971. [PMID: 28701379 DOI: 10.1073/pnas.1701137114] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) peptide deposition in brain parenchyma as plaques and in cerebral blood vessels as cerebral amyloid angiopathy (CAA). CAA deposition leads to several clinical complications, including intracerebral hemorrhage. The underlying molecular mechanisms that regulate plaque and CAA deposition in the vast majority of sporadic AD patients remain unclear. The clusterin (CLU) gene is genetically associated with AD and CLU has been shown to alter aggregation, toxicity, and blood-brain barrier transport of Aβ, suggesting it might play a key role in regulating the balance between Aβ deposition and clearance in both brain and blood vessels. Here, we investigated the effect of CLU on Aβ pathology using the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD amyloidosis on a Clu+/+ or Clu-/- background. We found a marked decrease in plaque deposition in the brain parenchyma but an equally striking increase in CAA within the cerebrovasculature of APP/PS1;Clu-/- mice. Surprisingly, despite the several-fold increase in CAA levels, APP/PS1;Clu-/- mice had significantly less hemorrhage and inflammation. Mice lacking CLU had impaired clearance of Aβ in vivo and exogenously added CLU significantly prevented Aβ binding to isolated vessels ex vivo. These findings suggest that in the absence of CLU, Aβ clearance shifts to perivascular drainage pathways, resulting in fewer parenchymal plaques but more CAA because of loss of CLU chaperone activity, complicating the potential therapeutic targeting of CLU for AD.
Collapse
|
39
|
Matukumalli SR, Tangirala R, Rao CM. Clusterin: full-length protein and one of its chains show opposing effects on cellular lipid accumulation. Sci Rep 2017; 7:41235. [PMID: 28120874 PMCID: PMC5264606 DOI: 10.1038/srep41235] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/16/2016] [Indexed: 12/31/2022] Open
Abstract
Proteins, made up of either single or multiple chains, are designed to carry out specific biological functions. We found an interesting example of a two-chain protein where administration of one of its chains leads to a diametrically opposite outcome than that reported for the full-length protein. Clusterin is a highly glycosylated protein consisting of two chains, α- and β-clusterin. We have investigated the conformational features, cellular localization, lipid accumulation, in vivo effects and histological changes upon administration of recombinant individual chains of clusterin. We demonstrate that recombinant α- and β-chains exhibit structural and functional differences and differ in their sub-cellular localization. Full-length clusterin is known to lower lipid levels. In contrast, we find that β-chain-treated cells accumulate 2-fold more lipid than controls. Interestingly, α-chain-treated cells do not show such increase. Rabbits injected with β-chain, but not α-chain, show ~40% increase in weight, with adipocyte hypertrophy, liver and kidney steatosis. Many, sometimes contrasting, roles are ascribed to clusterin in obesity, metabolic syndrome and related conditions. Our findings of differential localization and activities of individual chains of clusterin should help in understanding better the roles of clusterin in metabolism.
Collapse
Affiliation(s)
| | | | - C. M. Rao
- CSIR- Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| |
Collapse
|
40
|
Abstract
INTRODUCTION Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone, normally secreted from cells, that is up-regulated in Alzheimer disease and in many cancers. It plays important roles in protein homeostasis/proteostasis, inhibition of cell death pathways, and modulation of pro-survival signalling and transcriptional networks. Changes in the CLU gene locus are highly associated with Alzheimer disease, and many therapy-resistant cancers over-express CLU. The extensive post-translational processing and heterogeneous oligomerization of CLU have so far prevented any definitive structure determination. This in turn has meant that targeting CLU with small molecule inhibitors is challenging. Therefore, inhibiting CLU at the gene-expression level using siRNA or antisense is a valid approach to inhibit its function. Areas covered: This article reviews recent advances regarding the role of CLU in proteostasis, cellular trafficking, human diseases, and signalling pathways involved in oncogenesis. It addresses the rationale for CLU as a therapeutic target in cancer, and the current status of pre-clinical and clinical studies using CLU antisense inhibitor OGX011. Expert opinion: Discusses challenges facing the therapeutic targeting of CLU including rapid changes in the treatment landscape for prostate cancer with multiple new FDA approved drugs, selection of windows of intervention, and potential side effects when silencing CLU expression.
Collapse
Affiliation(s)
- Mark R Wilson
- a School of Biological Sciences , University of Wollongong , Wollongong , Australia
| | - Amina Zoubeidi
- b Department of Urologic Sciences, Vancouver Prostate Centre , University of British Columbia and Vancouver General Hospital , Vancouver , Canada
| |
Collapse
|
41
|
Fini ME, Bauskar A, Jeong S, Wilson MR. Clusterin in the eye: An old dog with new tricks at the ocular surface. Exp Eye Res 2016; 147:57-71. [PMID: 27131907 DOI: 10.1016/j.exer.2016.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 12/30/2022]
Abstract
The multifunctional protein clusterin (CLU) was first described in 1983 as a secreted glycoprotein present in ram rete testis fluid that enhanced aggregation ('clustering') of a variety of cells in vitro. It was also independently discovered in a number of other systems. By the early 1990s, CLU was known under many names and its expression had been demonstrated throughout the body, including in the eye. Its homeostatic activities in proteostasis, cytoprotection, and anti-inflammation have been well documented, however its roles in health and disease are still not well understood. CLU is prominent at fluid-tissue interfaces, and in 1996 it was demonstrated to be the most highly expressed transcript in the human cornea, the protein product being localized to the apical layers of the mucosal epithelia of the cornea and conjunctiva. CLU protein is also present in human tears. Using a preclinical mouse model for desiccating stress that mimics human dry eye disease, the authors recently demonstrated that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration in the tears. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to LGALS3 (galectin-3), a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. CLU depletion from the ocular surface epithelia is seen in a variety of inflammatory conditions in humans and mice that lead to squamous metaplasia and a keratinized epithelium. This suggests that CLU might have a specific role in maintaining mucosal epithelial differentiation, an idea that can now be tested using the mouse model for desiccating stress. Most excitingly, the new findings suggest that CLU could serve as a novel biotherapeutic for dry eye disease.
Collapse
Affiliation(s)
- M Elizabeth Fini
- USC Institute for Genetic Medicine and Departments of Cell & Neurobiology and Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Aditi Bauskar
- USC Institute for Genetic Medicine and Graduate Program in Medical Biology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Shinwu Jeong
- USC Institute for Genetic Medicine and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, New South Wales, 2522 Australia.
| |
Collapse
|
42
|
Vanhooren V, Navarrete Santos A, Voutetakis K, Petropoulos I, Libert C, Simm A, Gonos ES, Friguet B. Protein modification and maintenance systems as biomarkers of ageing. Mech Ageing Dev 2015; 151:71-84. [PMID: 25846863 DOI: 10.1016/j.mad.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 12/22/2022]
|
43
|
Rull A, Ordóñez-Llanos J, Sánchez-Quesada JL. The role of LDL-bound apoJ in the development of atherosclerosis. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
44
|
Rull A, Martínez-Bujidos M, Pérez-Cuellar M, Pérez A, Ordóñez-Llanos J, Sánchez-Quesada JL. Increased concentration of clusterin/apolipoprotein J (apoJ) in hyperlipemic serum is paradoxically associated with decreased apoJ content in lipoproteins. Atherosclerosis 2015; 241:463-70. [PMID: 26081122 DOI: 10.1016/j.atherosclerosis.2015.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/31/2015] [Accepted: 06/01/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Clusterin/apolipoprotein J (apoJ) circulates in blood in part associated to lipoproteins or in unbound form. When bound to HDL, apoJ is antiatherogenic by inhibiting endothelial cell apoptosis; thus, any factor modifying apoJ association to HDL would decrease its antiatherogenic function. However, the exact distribution of apoJ in each lipoprotein fraction, or in lipoprotein-non bound form has not been specifically investigated either in normolipemia or in dyslipemia. METHODS Basic lipid profile and apoJ concentration were determined in sera from 70 subjects, including a wide range of cholesterol and triglyceride concentrations. Lipoproteins were isolated by ultracentrifugation and their lipid and apolipoprotein composition was assessed. RESULTS In the overall population, serum apoJ positively associated with cholesterol, triglyceride and VLDL-C concentrations, and HDL-C and triglyceride were independent predictors of increased apoJ concentration. Approximately, 20.5% of circulating apoJ was associated with lipoproteins (18.5% HDL, 0.9% LDL and 1.1% VLDL) and 79.5% was not bound to lipoproteins. Serum apoJ concentration was higher in hypercholesterolemic (HC), hypertriglyceridemic (HTG) and combined hyperlipidemic (CHL) sera compared to normolipemic (NL) sera (HC, 98.15 ± 33.6 mg/L; HTG, 103.3 ± 36.8 mg/L; CHL, 131.7 ± 26.8 mg/L; NL, 66.7 ± 33.8 mg/L; P < 0.001). ApoJ distribution was also altered in hyperlipidemia; approximately 30% of circulating apoJ was associated to lipoproteins in the NL group whereas this proportion rounded 15% in hyperlipidemic subjects. CONCLUSIONS Our findings indicate that hyperlipidemia increases the concentration of apoJ in serum but, in turn, the content of lipoprotein-associated apoJ decreases. The redistribution of apoJ in hyperlipidemia could compromise the antiatherogenic properties of HDL.
Collapse
Affiliation(s)
- Anna Rull
- Cardiovascular Biochemistry Group, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Maria Martínez-Bujidos
- Cardiovascular Biochemistry Group, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain; Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Spain
| | - Montserrat Pérez-Cuellar
- Cardiovascular Biochemistry Group, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Antonio Pérez
- Endocrinology and Nutrition Department, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Jordi Ordóñez-Llanos
- Cardiovascular Biochemistry Group, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain; Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain.
| |
Collapse
|
45
|
Magnetic Bead-Based Serum Peptidome Profiling in Patients with Gestational Diabetes Mellitus. BIOMED RESEARCH INTERNATIONAL 2015; 2015:586309. [PMID: 26090425 PMCID: PMC4450277 DOI: 10.1155/2015/586309] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/06/2014] [Accepted: 08/28/2014] [Indexed: 12/18/2022]
Abstract
Gestational diabetes mellitus (GDM) is a frequent medical condition during pregnancy. Early diagnosis and treatment of GDM are crucial for both the mother and the baby. In the present study, we aimed to identify specific biomarkers to assist in the early detection of GDM and give some clues to the possible causes of GDM by comparing serum peptide profile differences between GDM patients and healthy controls. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was used in combination with weak cation exchange magnetic bead (WCX-MB). Levels of four peptides (4418.9, 2219.7, 2211.5, and 1533.4 Da) were significantly different. Interestingly, three of them (4418.9, 2211.5, and 1533.4 Da) were identified when GDM patients with two degrees of glucose intolerance were compared. Additionally, peptides 2211.5 and 1533.4 Da showed a decreasing trend as glucose intolerance increased, while peptide 4418.9 Da exhibited the reverse tendency. In conclusion, our study provides novel insights into the altered serum peptide profile of GDM patients. The specific candidate biomarkers may contribute to the development of GDM.
Collapse
|
46
|
Kontush A, Lindahl M, Lhomme M, Calabresi L, Chapman MJ, Davidson WS. Structure of HDL: particle subclasses and molecular components. Handb Exp Pharmacol 2015; 224:3-51. [PMID: 25522985 DOI: 10.1007/978-3-319-09665-0_1] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A molecular understanding of high-density lipoprotein (HDL) will allow a more complete grasp of its interactions with key plasma remodelling factors and with cell-surface proteins that mediate HDL assembly and clearance. However, these particles are notoriously heterogeneous in terms of almost every physical, chemical and biological property. Furthermore, HDL particles have not lent themselves to high-resolution structural study through mainstream techniques like nuclear magnetic resonance and X-ray crystallography; investigators have therefore had to use a series of lower resolution methods to derive a general structural understanding of these enigmatic particles. This chapter reviews current knowledge of the composition, structure and heterogeneity of human plasma HDL. The multifaceted composition of the HDL proteome, the multiple major protein isoforms involving translational and posttranslational modifications, the rapidly expanding knowledge of the HDL lipidome, the highly complex world of HDL subclasses and putative models of HDL particle structure are extensively discussed. A brief history of structural studies of both plasma-derived and recombinant forms of HDL is presented with a focus on detailed structural models that have been derived from a range of techniques spanning mass spectrometry to molecular dynamics.
Collapse
Affiliation(s)
- Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMR-ICAN 1166, Paris, France,
| | | | | | | | | | | |
Collapse
|
47
|
Martínez‐Bujidos M, Rull A, González‐Cura B, Pérez‐Cuéllar M, Montoliu‐Gaya L, Villegas S, Ordóñez‐Llanos J, Sánchez‐Quesada JL. Clusterin/apolipoprotein J binds to aggregated LDL in human plasma and plays a protective role against LDL aggregation. FASEB J 2014; 29:1688-700. [DOI: 10.1096/fj.14-264036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/01/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Maria Martínez‐Bujidos
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
- Biochemistry and Molecular Biology DepartmentUniversitat Autònoma de BarcelonaCerdanyolaSpain
| | - Anna Rull
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
| | - Beatriz González‐Cura
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
| | - Montserrat Pérez‐Cuéllar
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
| | - Laia Montoliu‐Gaya
- Protein Folding and Stability Group, Biochemistry and Molecular Biology DepartmentUniversitat Autònoma de BarcelonaCerdanyolaSpain
| | - Sandra Villegas
- Protein Folding and Stability Group, Biochemistry and Molecular Biology DepartmentUniversitat Autònoma de BarcelonaCerdanyolaSpain
| | - Jordi Ordóñez‐Llanos
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
- Biochemistry and Molecular Biology DepartmentUniversitat Autònoma de BarcelonaCerdanyolaSpain
| | - José Luis Sánchez‐Quesada
- Cardiovascular Biochemistry GroupResearch Institute of the Hospital de Sant Pau (IIB Sant Pau)BarcelonaSpain
| |
Collapse
|
48
|
Braesch-Andersen S, Beckman L, Paulie S, Kumagai-Braesch M. ApoD mediates binding of HDL to LDL and to growing T24 carcinoma. PLoS One 2014; 9:e115180. [PMID: 25513803 PMCID: PMC4267786 DOI: 10.1371/journal.pone.0115180] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022] Open
Abstract
Apolipoprotein (Apo) D is an important protein produced in many parts of the body. It is necessary for the development and repair of the brain and protection from oxidative stress. The purpose of this study was to investigate the extent to which apoD interacts with lipoproteins in human plasma. By using detergent-free ELISA, we show that immobilized monoclonal antibodies against apoD very efficiently bind to low density lipoprotein (LDL) from plasma; this binding is as equally efficient as binding to an anti-apoB monoclonal antibody. Adding detergent to the plasma inhibited the binding, suggesting that the binding is dependent on the presence of intact lipoprotein particles. Reversing the system by using immobilized anti-apoB revealed that the affinity of apoD for LDL is rather low, suggesting that multiple bindings are needed for a durable connection. Biosensor experiments using purified lipoproteins also showed that purified apoD and high density lipoprotein 3 (HDL3), a lipoprotein fraction rich in apoD, were both able to bind LDL very efficiently, indicating that the HDL3-LDL interaction may be a physiological consequence of the affinity of apoD for LDL. Furthermore, we found that apoD increases the binding of HDL to actively growing T24 bladder carcinoma cells but not to quiescent, contact-inhibited, confluent T24 cells. This result is especially intriguing given that the T24 supernatant only contained detectable levels of apoD after growth inhibition, raising the possibility that alternating the expression of apoD and a putative apoD-receptor could give direction to the flow of lipids. In the current paper, we conclude that apoD mediates binding of HDL to LDL and to growing T24 carcinomas, thereby highlighting the importance of apoD in lipid metabolism.
Collapse
Affiliation(s)
| | - Lena Beckman
- Mabtech, Nacka Strand, Sweden
- Metabolism Unit, Department of Endocrinology, Metabolism and Diabetes, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | |
Collapse
|
49
|
Afanasyeva MA, Britanova LV, Korneev KV, Mitkin NA, Kuchmiy AA, Kuprash DV. Clusterin is a potential lymphotoxin beta receptor target that is upregulated and accumulates in germinal centers of mouse spleen during immune response. PLoS One 2014; 9:e98349. [PMID: 24865838 PMCID: PMC4035297 DOI: 10.1371/journal.pone.0098349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/01/2014] [Indexed: 12/13/2022] Open
Abstract
Clusterin is a multifunctional protein that participates in tissue remodeling, apoptosis, lipid transport, complement-mediated cell lysis and serves as an extracellular chaperone. The role of clusterin in cancer and neurodegeneration has been extensively studied, however little is known about its functions in the immune system. Using expression profiling we found that clusterin mRNA is considerably down-regulated in mouse spleen stroma upon knock-out of lymphotoxin β receptor which plays pivotal role in secondary lymphoid organ development, maintenance and function. Using immunohistochemistry and western blot we studied clusterin protein level and distribution in mouse spleen and mesenteric lymph nodes in steady state and upon immunization with sheep red blood cells. We showed that clusterin protein, represented mainly by the secreted heterodimeric form, is present in all stromal compartments of secondary lymphoid organs except for marginal reticular cells. Clusterin protein level rose after immunization and accumulated in light zones of germinal centers in spleen--the effect that was not observed in lymph nodes. Regulation of clusterin expression by the lymphotoxin beta signaling pathway and its protein dynamics during immune response suggest a specific role of this enigmatic protein in the immune system that needs further study.
Collapse
Affiliation(s)
- Marina A. Afanasyeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Liudmila V. Britanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Kirill V. Korneev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Nikita A. Mitkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna A. Kuchmiy
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V. Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- * E-mail:
| |
Collapse
|
50
|
Yanni AE, Agrogiannis G, Gkekas C, Perrea D. Clusterin/Apolipoprotein J immunolocalization on carotid artery is affected by TNF-alpha, cigarette smoking and anti-platelet treatment. Lipids Health Dis 2014; 13:70. [PMID: 24758255 PMCID: PMC4005404 DOI: 10.1186/1476-511x-13-70] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/14/2014] [Indexed: 01/10/2023] Open
Abstract
Background Clusterin (CLU) /Apolipoprotein J is a protein biosensor of oxidative stress and inflammation, which is upregulated in many pathological processes including atherosclerosis. Previous studies have shown that in aortic tissue, CLU expression increases with atherosclerotic lesion progression and it has been coupled with vascular damage and coronary artery disease. A few studies enter into CLU and carotid atherosclerosis while the apolipoprotein’s expression on human carotid tissue and its association with parameters related to the disease development has not been examined. The present study was designed to reveal the relationships between the degree of CLU immunolocalization on carotid artery and demographic characteristics, blood parameters and pharmacological treatment of patients underwent internal carotid artery endarterectomy. Methods CLU expression was detected by immunohistochemistry in 42 carotid endarterectomy specimens. Patients’ serum levels of tumor necrosis factor-a (TNF-a), interleukin-6 (IL-6), high sensitive C-reactive protein (hsCRP) and classical parameters related to atherosclerosis such as lipid profile, as well as thrombosis related parameters such as fibrinogen, antithrombin III, protein C and protein S were determined. Demographic characteristics, smoking habits and the use of medications were recorded. Comparisons between groups were performed by students’t-test and analysis of variance. Independent associations with CLU expression on carotid tissue were denoted by linear regression analysis. Results CLU imuunolocalization was denser in smokers than in non-smokers (p = 0.041) while it was rarefied in specimens of patients on cropidogrel treatment (p = 0.045) compared to the rest not taking this medication. Clopidogrel intake was independent predictor of lower CLU expression on carotid artery (p =0.045). CLU was positively correlated with serum TNF-a concentration (r = 0.33, p = 0.040) that was independent predictor of higher expression of the apolipoprotein (p = 0.001). IL-6, hsCRP and classical parameters related to atherosclerosis and thrombosis were not associated with CLU immunolocalization. Conclusion Our study suggests that CLU expression on carotid artery is affected by TNF-alpha, cigarette smoking confirming its association with oxidative and cellular stress and anti-platelet medication reflecting the protective effects of such pharmacological treatment on vascular wall.
Collapse
Affiliation(s)
- Amalia E Yanni
- Department of Nutrition and Dietetics, Harokopio University of Athens, 70 El Venizelou Ave, Athens, Greece.
| | | | | | | |
Collapse
|