1
|
Fernández Encinas L, Lluch N, Wu AHB, Kaski JC, Badimon L, Cubedo J. A Novel ELISA for the Quantification of Serum Levels of 2 Glycosylated Variants of Apolipoprotein J: Biomarkers for Myocardial Ischemia. J Appl Lab Med 2023; 8:917-930. [PMID: 37473435 DOI: 10.1093/jalm/jfad034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/27/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Previous studies have pointed out a potential role of ApoJ-Glyc as a biomarker of cardiac ischemia. The aim of this study was to validate the analytical performance of 2 novel ELISAs against 2 different glycosylated ApoJ variants, ApoJ-GlycA2 and ApoJ-GlycA6. METHODS The analytical measuring range, limit of blank (LoB), lower limit of quantification (LoQ), precision, accuracy, recovery, cross-reactivity, and stability were evaluated in serum samples. RESULTS The analytical measuring range was 500-16 000 ng/mL for ApoJ-GlycA2 and 125-4000 ng/mL for ApoJ-GlycA6, with a LoB of 455 ng/mL and 121 ng/mL for ApoJ-GlycA2 and ApoJ-GlycA6, respectively. The LoQ was 500 ng/mL for ApoJ-GlycA2 and 125 ng/mL for ApoJ-GlycA6. The assay performance fulfills the acceptance criteria established in the European Medicines Agency Guideline on bioanalytical method validation. Specifically, the calibration range variability is <15% for ApoJ-GlycA2 and ApoJ-GlycA6; the accuracy is <15% for ApoJ-GlycA2 and ApoJ-GlycA6; the between- and within-run precision is <15% for ApoJ-GlycA6 and ≤20% for ApoJ-GlycA2; and the total allowable error is <30% for ApoJ-GlycA2 and ApoJ-GlycA6. Cross-reactivity studies revealed the absence of cross-reactivity with endogenous components of the matrix (using ApoJ-depleted serum), with nonglycosylated ApoJ and with transferrin (as a high abundant N-glycosylated serum protein). Both ApoJ-GlycA2 and ApoJ-GlycA6 measurements were stable after storage of serum samples at -80°C for 24 months. CONCLUSIONS The newly developed ELISAs to quantify ApoJ-GlycA2 and ApoJ-GlycA6 serum levels showed an acceptable analytical performance according to European Medicines Agency guidelines on bioanalytical method validation in terms of precision, accuracy, recovery, cross-reactivity, and stability.
Collapse
Affiliation(s)
| | - Nuria Lluch
- R&D department, GlyCardial Diagnostics, S.L., Barcelona, Spain
| | - Alan H B Wu
- Clinical Chemistry and Toxicology Laboratories, San Francisco General Hospital and Department of Laboratory Medicine, University of California, San Francisco, CA, United States
| | - Juan Carlos Kaski
- R&D department, GlyCardial Diagnostics, S.L., Barcelona, Spain
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, United Kingdom
| | - Lina Badimon
- R&D department, GlyCardial Diagnostics, S.L., Barcelona, Spain
- Cardiovascular Research Center-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Judit Cubedo
- R&D department, GlyCardial Diagnostics, S.L., Barcelona, Spain
| |
Collapse
|
2
|
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
|
3
|
Glycosylated clusterin species facilitate Aβ toxicity in human neurons. Sci Rep 2022; 12:18639. [PMID: 36329114 PMCID: PMC9633591 DOI: 10.1038/s41598-022-23167-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Clusterin (CLU) is one of the most significant genetic risk factors for late onset Alzheimer's disease (AD). However, the mechanisms by which CLU contributes to AD development and pathogenesis remain unclear. Studies have demonstrated that the trafficking and localisation of glycosylated CLU proteins is altered by CLU-AD mutations and amyloid-β (Aβ), which may contribute to AD pathogenesis. However, the roles of non-glycosylated and glycosylated CLU proteins in mediating Aβ toxicity have not been studied in human neurons. iPSCs with altered CLU trafficking were generated following the removal of CLU exon 2 by CRISPR/Cas9 gene editing. Neurons were generated from control (CTR) and exon 2 -/- edited iPSCs and were incubated with aggregated Aβ peptides. Aβ induced changes in cell death and neurite length were quantified to determine if altered CLU protein trafficking influenced neuronal sensitivity to Aβ. Finally, RNA-Seq analysis was performed to identify key transcriptomic differences between CLU exon 2 -/- and CTR neurons. The removal of CLU exon 2, and the endoplasmic reticulum (ER)-signal peptide located within, abolished the presence of glycosylated CLU and increased the abundance of intracellular, non-glycosylated CLU. While non-glycosylated CLU levels were unaltered by Aβ25-35 treatment, the trafficking of glycosylated CLU was altered in control but not exon 2 -/- neurons. The latter also displayed partial protection against Aβ-induced cell death and neurite retraction. Transcriptome analysis identified downregulation of multiple extracellular matrix (ECM) related genes in exon 2 -/- neurons, potentially contributing to their reduced sensitivity to Aβ toxicity. This study identifies a crucial role of glycosylated CLU in facilitating Aβ toxicity in human neurons. The loss of these proteins reduced both, cell death and neurite damage, two key consequences of Aβ toxicity identified in the AD brain. Strikingly, transcriptomic differences between exon 2 -/- and control neurons were small, but a significant and consistent downregulation of ECM genes and pathways was identified in exon 2 -/- neurons. This may contribute to the reduced sensitivity of these neurons to Aβ, providing new mechanistic insights into Aβ pathologies and therapeutic targets for AD.
Collapse
|
4
|
Construction of a Colorectal Cancer Prognostic Risk Model and Screening of Prognostic Risk Genes Using Machine-Learning Algorithms. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9408839. [PMID: 36267311 PMCID: PMC9578894 DOI: 10.1155/2022/9408839] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 12/09/2022]
Abstract
This study is aimed at constructing a prognostic risk model for colorectal cancer (CRC) using machine-learning algorithms to provide accurate staging and screening of credible prognostic risk genes. We extracted CRC data from GSE126092 and GSE156355 of the Gene Expression Omnibus (GEO) database and datasets from TCGA to analyze the differentially expressed genes (DEGs) using bioinformatics analysis. Among the 330 shared DEGs related to CRC prognosis, we divided the analysis period into different phases and applied univariate COX regression, LASSO, and multivariate COX regression analysis. GO analysis and KEGG analysis revealed that the functions of these DEGs were primarily focused on cell cycle, DNA replication, cell mitosis, and other related functions, and this confirmed our results from a biological perspective. Finally, a prognostic risk model for CRC based on the CHGA, CLU, PLK1, AXIN2, NR3C2, IL17RB, GCG, and AJUBA genes was constructed, and the risk score enabled us to predict the prognosis for CRC. To obtain a comprehensive and accurate model, we used both internal and external evaluations, and the model was able to correctly differentiate patients with CRC into a high-risk group with poor prognosis and a low-risk group with good prognosis. The AUC values of the 3-, 5-, and 10-year survival ROC curves were 0.715, 0.721, and 0.777, respectively, according to the internal evaluation, and the AUC values were 0.606, 0.698, and 0.608, respectively, for the external evaluation using GSE39582 from the GEO database. We determined that CLU, PLK1, and IL17RB could be considered to be independent prognostic factors for CRC with significantly different expression (P < 0.05). Using machine-learning methods, a prognostic risk model comprised of eight genes was constructed. Not only does this model provide improved treatment guidance, but it also provides a novel perspective for analyzing survival conditions at a deeper biological level.
Collapse
|
5
|
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
|
6
|
Andújar-Vera F, García-Fontana C, Sanabria-de la Torre R, González-Salvatierra S, Martínez-Heredia L, Iglesias-Baena I, Muñoz-Torres M, García-Fontana B. Identification of Potential Targets Linked to the Cardiovascular/Alzheimer's Axis through Bioinformatics Approaches. Biomedicines 2022; 10:389. [PMID: 35203598 PMCID: PMC8962298 DOI: 10.3390/biomedicines10020389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 12/23/2022] Open
Abstract
The identification of common targets in Alzheimer's disease (AD) and cardiovascular disease (CVD) in recent years makes the study of the CVD/AD axis a research topic of great interest. Besides aging, other links between CVD and AD have been described, suggesting the existence of common molecular mechanisms. Our study aimed to identify common targets in the CVD/AD axis. For this purpose, genomic data from calcified and healthy femoral artery samples were used to identify differentially expressed genes (DEGs), which were used to generate a protein-protein interaction network, where a module related to AD was identified. This module was enriched with the functionally closest proteins and analyzed using different centrality algorithms to determine the main targets in the CVD/AD axis. Validation was performed by proteomic and data mining analyses. The proteins identified with an important role in both pathologies were apolipoprotein E and haptoglobin as DEGs, with a fold change about +2 and -2, in calcified femoral artery vs healthy artery, respectively, and clusterin and alpha-2-macroglobulin as close interactors that matched in our proteomic analysis. However, further studies are needed to elucidate the specific role of these proteins, and to evaluate its function as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Francisco Andújar-Vera
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Raquel Sanabria-de la Torre
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Sheila González-Salvatierra
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Iván Iglesias-Baena
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain;
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
7
|
Wittwer J, Bradley D. Clusterin and Its Role in Insulin Resistance and the Cardiometabolic Syndrome. Front Immunol 2021; 12:612496. [PMID: 33717095 PMCID: PMC7946829 DOI: 10.3389/fimmu.2021.612496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The cardiometabolic syndrome involves a clustering of metabolic and cardiovascular factors which increase the risk of patients developing both Type 2 Diabetes Mellitus and cardio/cerebrovascular disease. Although the mechanistic underpinnings of this link remain uncertain, key factors include insulin resistance, excess visceral adiposity, atherogenic dyslipidemia, and endothelial dysfunction. Of these, a state of resistance to insulin action in overweight/obese patients appears to be central to the pathophysiologic process. Given the increasing prevalence of obesity-related Type 2 Diabetes, coupled with the fact that cardiovascular disease is the number one cause of mortality in this patient population, a more thorough understanding of the cardiometabolic syndrome and potential options to mitigate its risk is imperative. Inherent in the pathogenesis of insulin resistance is an underlying state of chronic inflammation, at least partly in response to excess adiposity. Within obese adipose tissue, an immunomodulatory shift occurs, involving a preponderance of pro-inflammatory immune cells and cytokines/adipokines, along with antigen presentation by adipocytes. Therefore, various adipokines differentially expressed by obese adipocytes may have a significant effect on cardiometabolism. Clusterin is a molecular chaperone that is widely produced by many tissues throughout the body, but is also preferentially overexpressed by obese compared lean adipocytes and relates strongly to multiple components of the cardiometabolic syndrome. Herein, we summarize the known and potential roles of circulating and adipocyte-specific clusterin in cardiometabolism and discuss potential further investigations to determine if clusterin is a viable target to attenuate both metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer Wittwer
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
8
|
Praharaj PP, Patra S, Panigrahi DP, Patra SK, Bhutia SK. Clusterin as modulator of carcinogenesis: A potential avenue for targeted cancer therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188500. [PMID: 33385484 DOI: 10.1016/j.bbcan.2020.188500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
9
|
Geßner C, Stillger MN, Mölders N, Fabrizius A, Folkow LP, Burmester T. Cell Culture Experiments Reveal that High S100B and Clusterin Levels may Convey Hypoxia-tolerance to the Hooded Seal (Cystophora cristata) Brain. Neuroscience 2020; 451:226-239. [PMID: 33002555 DOI: 10.1016/j.neuroscience.2020.09.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
While the brain of most mammals suffers from irreversible damage after only short periods of low oxygen levels (hypoxia), marine mammals are excellent breath-hold divers that have adapted to hypoxia. In addition to physiological adaptations, such as large oxygen storing capacity and strict oxygen economy during diving, the neurons of the deep-diving hooded seal (Cystophora cristata) have an intrinsic tolerance to hypoxia. We aim to understand the molecular basis of this neuronal hypoxia tolerance. Previously, transcriptomics of the cortex of the hooded seal have revealed remarkably high expression levels of S100B and clusterin (apolipoprotein J) when compared to the ferret, a non-diving carnivore. Both genes have much-debated roles in hypoxia and oxidative stress. Here, we evaluated the effects of S100B and of two isoforms of clusterin (soluble and nucleus clusterin) on the survival, metabolic activity and the amount of reactive oxygen species (ROS) in HN33 neuronal mouse cells exposed to hypoxia and oxidative stress. S100B and soluble clusterin had neuroprotective effects, with reduced ROS-levels and retention of normoxic energy status of cells during both stress conditions. The protective effects of nucleus clusterin were restricted to hypoxia. S100B and clusterin showed purifying selection in marine and terrestrial mammals, indicating a functional conservation across species. Immunofluorescence revealed identical cellular distributions of S100B and clusterin in mice, ferrets and hooded seals, further supporting the functional conservation. Taken together, our data suggest that the neuroprotective effects of all three proteins are exclusively facilitated by their increased expression in the brain of the hooded seal.
Collapse
Affiliation(s)
- Cornelia Geßner
- Institute of Zoology, University of Hamburg, 20146 Hamburg, Germany.
| | | | - Naomi Mölders
- Institute of Zoology, University of Hamburg, 20146 Hamburg, Germany
| | - Andrej Fabrizius
- Institute of Zoology, University of Hamburg, 20146 Hamburg, Germany
| | - Lars P Folkow
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway
| | | |
Collapse
|
10
|
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: 242] [Impact Index Per Article: 40.3] [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
|
11
|
Tajbakhsh A, Pasdar A, Rezaee M, Fazeli M, Soleimanpour S, Hassanian SM, FarshchiyanYazdi Z, Younesi Rad T, Ferns GA, Avan A. The current status and perspectives regarding the clinical implication of intracellular calcium in breast cancer. J Cell Physiol 2018; 233:5623-5641. [PMID: 29150934 DOI: 10.1002/jcp.26277] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Calcium ions (Ca2+ ) act as second messengers in intracellular signaling. Ca2+ pumps, channels, sensors, and calcium binding proteins, regulate the concentrations of intracellular Ca2+ as a key regulator of important cellular processes such as gene expression, proliferation, differentiation, DNA repair, apoptosis, metastasis, and hormone secretion. Intracellular Ca2+ also influences the functions of several organelles, that include: the endoplasmic reticulum, mitochondria, the Golgi, and cell membrane both in normal and breast cancer cells. In breast cancer, the disruption of intracellular: Ca2+ homeostasis may cause tumor progression by affecting key factors/pathways including phospholipase C (PLC), inositol 1,4,5-trisphosphate (IP3), calmodulin (CaM), nuclear factor of activated T-cells (NFAT), calpain, calmodulin-dependent protein kinase II (CaMKII), mitogen-activated protein kinase (MAPK), epithelial-mesenchymal transition (EMT), vascular endothelial growth factor (VEGF), poly (ADP-Ribose) polymerase-1 (PARP1), estrogen, and estrogen receptor. Because the foregoing molecules play crucial roles in breast cancer, the factors/pathways influencing intracellular Ca2+ concentrations are putative targets for cancer treatment, using drugs such as Mephebrindole, Tilapia piscidin 4, Nifetepimine, Paricalcitol, and Prednisolone. We have explored the factors/pathways which are related to breast cancer and Ca2+ homeostasis and signaling in this review, and also discussed their potential as biomarkers for breast cancer staging, prognosis, and therapy.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Pasdar
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, UK.,Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rezaee
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Fazeli
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra FarshchiyanYazdi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Laboratory Sciences, Faculty of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tayebe Younesi Rad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Laboratory Sciences, Faculty of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, Sussex, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Protective effect of clusterin on rod photoreceptor in rat model of retinitis pigmentosa. PLoS One 2017; 12:e0182389. [PMID: 28767729 PMCID: PMC5540409 DOI: 10.1371/journal.pone.0182389] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Retinitis Pigmentosa (RP) begins with the death of rod photoreceptors and is slowly followed by a gradual loss of cones and a rearrangement of the remaining retinal neurons. Clusterin is a chaperone protein that protects cells and is involved in various pathophysiological stresses, including retinal degeneration. Using a well-established transgenic rat model of RP (rhodopsin S334ter), we investigated the effects of clusterin on rod photoreceptor survival. To investigate the role of clusterin in S334ter-line3 retinas, Voronoi analysis and immunohistochemistry were used to evaluate the geometry of rod distribution. Additionally, immunoblot analysis, Bax activation, STAT3 and Akt phosphorylation were used to evaluate the pathway involved in rod cell protection. In this study, clusterin (10μg/ml) intravitreal treatment produced robust preservation of rod photoreceptors in S334ter-line3 retina. The mean number of rods in 1mm2 was significantly greater in clusterin injected RP retinas (postnatal (P) 30, P45, P60, & P75) than in age-matched saline injected RP retinas (P<0.01). Clusterin activated Akt, STAT3 and significantly reduced Bax activity; in addition to inducing phosphorylated STAT3 in Müller cells, which suggests it may indirectly acts on photoreceptors. Thus, clusterin treatment may interferes with mechanisms leading to rod death by suppressing cell death through activation of Akt and STAT3, followed by Bax suppression. Novel insights into the pathway of how clusterin promotes the rod cell survival suggest this treatment may be a potential therapeutic strategy to slow progression of vision loss in human RP.
Collapse
|
13
|
Terenina E, Fabre S, Bonnet A, Monniaux D, Robert-Granié C, SanCristobal M, Sarry J, Vignoles F, Gondret F, Monget P, Tosser-Klopp G. Differentially expressed genes and gene networks involved in pig ovarian follicular atresia. Physiol Genomics 2017; 49:67-80. [DOI: 10.1152/physiolgenomics.00069.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/28/2016] [Accepted: 12/02/2016] [Indexed: 01/08/2023] Open
Abstract
Ovarian folliculogenesis corresponds to the development of follicles leading to either ovulation or degeneration, this latter process being called atresia. Even if atresia involves apoptosis, its mechanism is not well understood. The objective of this study was to analyze global gene expression in pig granulosa cells of ovarian follicles during atresia. The transcriptome analysis was performed on a 9,216 cDNA microarray to identify gene networks and candidate genes involved in pig ovarian follicular atresia. We found 1,684 significantly regulated genes to be differentially regulated between small healthy follicles and small atretic follicles. Among them, 287 genes had a fold-change higher than two between the two follicle groups. Eleven genes ( DKK3, GADD45A, CAMTA2, CCDC80, DAPK2, ECSIT, MSMB, NUPR1, RUNX2, SAMD4A, and ZNF628) having a fold-change higher than five between groups could likely serve as markers of follicular atresia. Moreover, automatic confrontation of deregulated genes with literature data highlighted 93 genes as regulatory candidates of pig granulosa cell atresia. Among these genes known to be inhibitors of apoptosis, stimulators of apoptosis, or tumor suppressors INHBB, HNF4, CLU, different interleukins ( IL5, IL24), TNF-associated receptor ( TNFR1), and cytochrome-c oxidase ( COX) were suggested as playing an important role in porcine atresia. The present study also enlists key upstream regulators in follicle atresia based on our results and on a literature review. The novel gene candidates and gene networks identified in the current study lead to a better understanding of the molecular regulation of ovarian follicular atresia.
Collapse
Affiliation(s)
- Elena Terenina
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Stephane Fabre
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Agnès Bonnet
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Danielle Monniaux
- INRA UMR 0085, CNRS UMR 7247, Université Francois Rabelais de Tours, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | | | - Magali SanCristobal
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Julien Sarry
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Florence Vignoles
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Florence Gondret
- INRA, UMR1348 Pegase, Saint‐Gilles, France; and
- AgroCampus-Ouest, UMR1348 Pegase, Saint‐Gilles, France
| | - Philippe Monget
- INRA UMR 0085, CNRS UMR 7247, Université Francois Rabelais de Tours, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | | |
Collapse
|
14
|
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
|
15
|
Hurwitz A, Ruutiainen-Altman K, Marzella L, Botero L, Dushnik M, Adashi EY. Follicular Atresia as an Apoptotic Process: Atresia-Associated Increase in the Ovarian Expression of the Putative Apoptotic Marker Sulfated Glycoprotein-2. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769600300407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | | | - Louis Marzella
- Division os Reproductive Endocrinology, Department of Obstetnes and Gynecology, and the Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Luis Botero
- Division os Reproductive Endocrinology, Department of Obstetnes and Gynecology, and the Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; Universidad Javeriana, Hospital San Ignacio, Departamento de Gineco-Obstetricia (3rd PISO), Bogota, Colombia
| | - Matat Dushnik
- Division os Reproductive Endocrinology, Department of Obstetnes and Gynecology, and the Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eli Y. Adashi
- Division os Reproductive Endocrinology, Department of Obstetnes and Gynecology, and the Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; Division of Reproductive Endocrinology, Departments of Obstetrics/Gynecology and Physiology, 405 W. Redwood Street, Third Floor, Baltimore, MD 21201
| |
Collapse
|
16
|
Fukuda T, Miyake H, Enatsu N, Matsushita K, Fujisawa M. Seminal level of clusterin in infertile men as a significant biomarker reflecting spermatogenesis. Andrologia 2016; 48:1188-1194. [DOI: 10.1111/and.12558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2016] [Indexed: 11/29/2022] Open
Affiliation(s)
- T. Fukuda
- Division of Urology; Kobe University Graduate School of Medicine; Kobe Japan
| | - H. Miyake
- Division of Urology; Kobe University Graduate School of Medicine; Kobe Japan
| | - N. Enatsu
- Division of Urology; Kobe University Graduate School of Medicine; Kobe Japan
| | - K. Matsushita
- Division of Urology; Kobe University Graduate School of Medicine; Kobe Japan
| | - M. Fujisawa
- Division of Urology; Kobe University Graduate School of Medicine; Kobe Japan
| |
Collapse
|
17
|
Ronquist G. Prostasomes: Their Characterisation: Implications for Human Reproduction: Prostasomes and Human Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 868:191-209. [PMID: 26178851 PMCID: PMC7120776 DOI: 10.1007/978-3-319-18881-2_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The prostate is a principal accessory genital gland that is vital for normal fertility. Epithelial cells lining the prostate acini release in a defined fashion (exocytosis) organellar nanosized structures named prostasomes. They are involved in the protection of sperm cells against immune response in the female reproductive tract by modulating the complement system and by inhibiting monocyte and neutrophil phagocytosis and lymphocyte proliferation. The immunomodulatory function most probably involves small non-coding RNAs present in prostasomes. Prostasomes have also been proposed to regulate the timing of sperm cell capacitation and induction of the acrosome reaction, since they are rich in various transferable bioactive molecules (e.g. receptors and enzymes) that promote the fertilising ability of sperm cells. Antigenicity of sperm cells has been well documented and implicated in involuntary immunological infertility of human couples, and antisperm antibodies (ASA) occur in several body fluids. The propensity of sperm cells to carry attached prostasomes suggests that they are a new category of sperm antigens. Circulating human ASA recognise prostasomes, and among 12 identified prostasomal antigens, prolactin- inducible protein (95 %) and clusterin (85 %) were immunodominant at the expense of the other 10 that were sporadically occurring.
Collapse
Affiliation(s)
- Gunnar Ronquist
- Department of Medical Sciences, Clinical Chemistry, The University of Uppsala, 751 85, Uppsala, Sweden,
| |
Collapse
|
18
|
Zhang SW, Shao DD, Zhang SY, Wang YB. Prioritization of candidate disease genes by enlarging the seed set and fusing information of the network topology and gene expression. MOLECULAR BIOSYSTEMS 2014; 10:1400-8. [PMID: 24695957 DOI: 10.1039/c3mb70588a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The identification of disease genes is very important not only to provide greater understanding of gene function and cellular mechanisms which drive human disease, but also to enhance human disease diagnosis and treatment. Recently, high-throughput techniques have been applied to detect dozens or even hundreds of candidate genes. However, experimental approaches to validate the many candidates are usually time-consuming, tedious and expensive, and sometimes lack reproducibility. Therefore, numerous theoretical and computational methods (e.g. network-based approaches) have been developed to prioritize candidate disease genes. Many network-based approaches implicitly utilize the observation that genes causing the same or similar diseases tend to correlate with each other in gene-protein relationship networks. Of these network approaches, the random walk with restart algorithm (RWR) is considered to be a state-of-the-art approach. To further improve the performance of RWR, we propose a novel method named ESFSC to identify disease-related genes, by enlarging the seed set according to the centrality of disease genes in a network and fusing information of the protein-protein interaction (PPI) network topological similarity and the gene expression correlation. The ESFSC algorithm restarts at all of the nodes in the seed set consisting of the known disease genes and their k-nearest neighbor nodes, then walks in the global network separately guided by the similarity transition matrix constructed with PPI network topological similarity properties and the correlational transition matrix constructed with the gene expression profiles. As a result, all the genes in the network are ranked by weighted fusing the above results of the RWR guided by two types of transition matrices. Comprehensive simulation results of the 10 diseases with 97 known disease genes collected from the Online Mendelian Inheritance in Man (OMIM) database show that ESFSC outperforms existing methods for prioritizing candidate disease genes. The top prediction results of Alzheimer's disease are consistent with previous literature reports.
Collapse
Affiliation(s)
- Shao-Wu Zhang
- College of Automation, Northwestern Polytechnical University, 710072, Xi'an, China.
| | | | | | | |
Collapse
|
19
|
Park S, Mathis KW, Lee IK. The physiological roles of apolipoprotein J/clusterin in metabolic and cardiovascular diseases. Rev Endocr Metab Disord 2014; 15:45-53. [PMID: 24097125 DOI: 10.1007/s11154-013-9275-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Several isoforms of apolipoprotein J/clusterin (CLU) are encoded from a single gene located on chromosome 8 in humans. These isoforms are ubiquitously expressed in the tissues, and have been implicated in aging, neurodegenerative disorders, cancer progression, and metabolic/cardiovascular diseases including dyslipidemia, diabetes, atherosclerosis and myocardial infarction. The conventional secreted form of CLU (sCLU) is thought to be a component of high density lipoprotein-cholesterol. sCLU functions as a chaperone for misfolded proteins and it is thought to promote survival by reducing oxidative stress. Nuclear CLU, a truncated CLU formed by alternative splicing, is responsible for promoting apoptosis via a Bax-dependent pathway. There are putative regulatory sites in the promoter regions of CLU, which are occupied by transcription factors such as transforming growth factor (TGF)-β inhibitory element, activator protein-1, CLU-specific elements, and carbohydrate response element. However, the molecular mechanisms underlying the distinct roles of CLU in a variety of conditions remain unclear. Although the function of CLU in cancer or neurological disease has been studied intensively for three decades, physiological roles of CLU seem unexplored in the cardiovascular system and metabolic diseases. In this review, we will discuss general characteristics and regulations of CLU based on previous literature and assess the recent findings associated with its physiological roles in different tissues including the vasculature, heart, liver, kidney, adipose tissue, and brain.
Collapse
Affiliation(s)
- S Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | | | | |
Collapse
|
20
|
Han Z, Wang Z, Cheng G, Liu B, Li P, Li J, Wang W, Yin C, Zhang W. Presence, localization, and origin of clusterin in normal human spermatozoa. J Assist Reprod Genet 2012; 29:751-7. [PMID: 22552734 DOI: 10.1007/s10815-012-9779-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 04/18/2012] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Clusterin in mammalian semen is a secretory form of clusterin (sCLU) with the heterodimeric structure. It is secreted by the epididymis and seminal vesicle. It is generally agreed that clusterin mainly exists on the surface of abnormal spermatozoa and is implicated in decreased sperm motility, sperm aggregation and infertility. However, few studies observe clusterin in normal spermatozoa, which is presumed to be a novel form. Up to now, the systematical information about the presence, localization, origin and function of clusterin in normal human spermatozoa has yet not been established. The aim of our current study is to systematically research clusterin in normal human spermatozoa. METHODS We detected the presence of clusterin via western blot, explored the localization of clusterin using immunofluorescence, and investigated the origin and distribution of clusterin in human testis by western blot and immunohistochemistry. RESULTS We found native clusterin in the inner plasma membrane of normal human spermatozoa. It was derived from the testis and showed similar molecular weight and heterodimeric structure compared with sCLU in semen and on the surface of abnormal spermatozoa. CONCLUSION Clusterin in normal spermatozoa should be self-synthesized during the later stage of spermatogenesis. The different localization and origin suggested that the clusterin observed by us may be a novel form compared with conventional sCLU on the surface of abnormal spermatozoa.
Collapse
Affiliation(s)
- Zhijian Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Analysis of dynamic changes in the proteome of a Bcl-XL overexpressing Chinese hamster ovary cell culture during exponential and stationary phases. Biotechnol Prog 2012; 28:814-23. [DOI: 10.1002/btpr.1534] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/23/2012] [Indexed: 11/07/2022]
|
22
|
Abstract
Clusterin, also known as apolipoprotein J, is a ubiquitous multifunctional glycoprotein. Following its identification in 1983, clusterin was found to be clearly increased in Alzheimer's disease (AD). Later research demonstrated that clusterin could bind amyloid-beta (Abeta) peptides and prevent fibril formation, a hallmark of AD pathology. In addition to preventing excessive inflammation, intracellular clusterin was found to reduce apoptosis and oxidative stress. Although early studies were inconclusive, two recent large-scale genome-wide association studies (GWAS) independently identified variants within the clusterin gene as risk factors for developing AD. This review focuses on the characteristics of clusterin and possible mechanisms of its relationship to AD.
Collapse
Affiliation(s)
- Zhong-Chen Wu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | | | | | | |
Collapse
|
23
|
Guerreiro RJ, Gustafson DR, Hardy J. The genetic architecture of Alzheimer's disease: beyond APP, PSENs and APOE. Neurobiol Aging 2012; 33:437-56. [PMID: 20594621 PMCID: PMC2980860 DOI: 10.1016/j.neurobiolaging.2010.03.025] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 03/01/2010] [Accepted: 03/11/2010] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a complex disorder with a clear genetic component. Three genes have been identified as the cause of early onset familial AD (EOAD). The most common form of the disease, late onset Alzheimer's disease (LOAD), is, however, a sporadic one presenting itself in later stages of life. The genetic component of this late onset form of AD has been the target of a large number of studies, because only one genetic risk factor (APOE4) has been consistently associated with the disease. However, technological advances allow new approaches in the study of complex disorders. In this review, we discuss the new results produced by genome wide association studies, in light of the current knowledge of the complexity of AD genetics.
Collapse
Affiliation(s)
- Rita J Guerreiro
- Laboratory of Neurogenetics, National Institute of Aging, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
24
|
Fjeldbo CS, Bakke I, Erlandsen SE, Holmseth J, Lægreid A, Sandvik AK, Thommesen L, Bruland T. Gastrin upregulates the prosurvival factor secretory clusterin in adenocarcinoma cells and in oxyntic mucosa of hypergastrinemic rats. Am J Physiol Gastrointest Liver Physiol 2012; 302:G21-33. [PMID: 21995960 DOI: 10.1152/ajpgi.00197.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We show that the gastric hormone gastrin induces the expression of the prosurvival secretory clusterin (sCLU) in rat adenocarcinoma cells. Clusterin mRNA was still upregulated in the presence of the protein synthesis inhibitor cycloheximide, although at a lower level. This indicates that gastrin induces clusterin transcription independently of de novo protein synthesis but requires de novo protein synthesis of signal transduction pathway components to achieve maximal expression level. Luciferase reporter assay indicates that the AP-1 transcription factor complex is involved in gastrin-mediated activation of the clusterin promoter. Gastrin-induced clusterin expression and subsequent secretion is dependent on sustained treatment, because removal of gastrin after 1-2 h abolished the response. Neutralization of secreted clusterin by a specific antibody abolished the antiapoptotic effect of gastrin on serum starvation-induced apoptosis, suggesting that extracellular clusterin is involved in gastrin-mediated inhibition of apoptosis. The clusterin response to gastrin was validated in vivo in hypergastrinemic rats, showing increased clusterin expression in the oxyntic mucosa, as well as higher levels of clusterin in plasma. In normal rat oxyntic mucosa, clusterin protein was strongly expressed in chromogranin A-immunoreactive neuroendocrine cells, of which the main cell type was the histidine decarboxylase-immunoreactive enterochromaffin-like (ECL) cell. The association of clusterin with neuroendocrine differentiation was further confirmed in human gastric ECL carcinoids. Interestingly, in hypergastrinemic rats, clusterin-immunoreactive cells formed distinct groups of diverse cells at the base of many glands. Our results suggest that clusterin may contribute to gastrin's growth-promoting effect on the oxyntic mucosa.
Collapse
Affiliation(s)
- Christina Sæten Fjeldbo
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim N, Han JY, Roh GS, Kim HJ, Kang SS, Cho GJ, Park JY, Choi WS. Nuclear clusterin is associated with neuronal apoptosis in the developing rat brain upon ethanol exposure. Alcohol Clin Exp Res 2011; 36:72-82. [PMID: 21762182 DOI: 10.1111/j.1530-0277.2011.01588.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is often accompanied by reduced brain volumes, reflecting brain cell death induced by ethanol, but the molecular mechanisms were less elucidated. This study was set up to investigate whether clusterin (Clu) was involved in neuronal cell death in developing rats. METHODS Seven-day-old rats were subcutaneously injected with 20% ethanol in normal saline at 3 g/kg twice. The upregulation of Clu and cell death was detected by immunohistochemistry, immunofluorescence microscopy, and/or Western blotting. Protein-protein interaction was detected by immunoprecipitation and immunoblotting. To identify the isoform interacting with Bcl-XL, HT22 mouse hippocampal cells were transfected with nuclear Clu(nClu)- or secretory Clu-expressing vector, and confocal microscopy was performed. Clu transcripts were knocked down in primary cortical cells using siRNA. RESULTS We found that Clu increased in the cerebral cortex following acute ethanol treatment. The Clu upregulation was related to increased cell death, which was assessed by activated caspase-3 and TUNEL staining. The upregulated Clu was a nuclear isoform that was nuclear translocated upon ethanol exposure and interacted with Bcl-XL, mediating apoptosis. CONCLUSIONS This study shows that nClu plays a pro-apoptotic role in ethanol-induced cell death in the developing brain, providing new insights for development of FASD.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Anatomy and Neurobiology, Health Science Institute, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Daimon M, Oizumi T, Karasawa S, Kaino W, Takase K, Tada K, Jimbu Y, Wada K, Kameda W, Susa S, Muramatsu M, Kubota I, Kawata S, Kato T. Association of the clusterin gene polymorphisms with type 2 diabetes mellitus. Metabolism 2011; 60:815-22. [PMID: 20850846 DOI: 10.1016/j.metabol.2010.07.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/26/2010] [Accepted: 07/26/2010] [Indexed: 12/21/2022]
Abstract
The association of the clusterin (CLU) gene polymorphism (single nucleotide polymorphisms [SNPs] 1-4: rs1532278, rs1532277, rs2279590, and rs2279591, respectively) with type 2 diabetes mellitus was examined using a population of the Funagata study (n [male-female] = 1631 [741:884]; age, 62.0 ± 12.1 years), a Japanese community-based study. Single nucleotide polymorphisms 1 to 3 were significantly associated with hemoglobin A(1c) levels (P = .0154, .0021, and .0006, respectively) and diabetes (.0310, .0170, and .0021, respectively). A case-control association study of SNP 3 with diabetes by multiple logistic regression analysis showed a significant association of genotype AA (the at-risk genotype) with an odds ratio (OR) of 2.33 (P = .0039) independently of age and sex. The association was marginally validated by a study with another Japanese community-based sample, the Takahata Study (n [male-female] = 2.948 [1333:1615]; age, 63.0 ± 10.2 years) (OR, 1.59; P = .0595; χ(2)P = .0264). When the 2 samples were combined, the association became more significant (OR, 1.75; P = .0025). In subjects with the non-at-risk genotypes, the insulin resistance index--homeostasis model assessment of insulin resistance (HOMA-R)--increased significantly (P < .0001) and the insulin secretion index--HOMA-β--appeared to decrease (P = .1803 and .0097, respectively, for the genotypes AG and GG) as the glucose tolerance progressed toward diabetes (normal glucose tolerance to glucose intolerance to diabetes). However, in subjects with the at-risk genotype, HOMA-R and HOMA-β showed a significant increase already in the subjects with normal glucose tolerance (P = .0239 and .0305, respectively); and as the glucose tolerance progressed toward diabetes, HOMA-R stayed approximately the same, whereas HOMA-β decreased significantly (P = .0332). The CLU gene was associated with diabetes, probably through an increase in insulin resistance primarily and through an impairment of insulin secretion secondarily.
Collapse
Affiliation(s)
- Makoto Daimon
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata 990-9585, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sherva R, Farrer LA. Power and pitfalls of the genome-wide association study approach to identify genes for Alzheimer's disease. Curr Psychiatry Rep 2011; 13:138-46. [PMID: 21312009 PMCID: PMC3154249 DOI: 10.1007/s11920-011-0184-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Until recently, the search for genes contributing to Alzheimer's disease (AD) had been slow and disappointing, with the notable exception of the APOE ε4 allele, which increases risk and reduces the age at onset of AD in a dose-dependent fashion. Findings from genome-wide association studies (GWAS) made up of fewer than several thousand cases and controls each have not been replicated. Efforts of several consortia--each assembling much larger datasets with sufficient power to detect loci conferring small changes in AD risk--have resulted in robust associations with many novel genes involved in multiple biological pathways. Complex data mining strategies are being used to identify additional members of these pathways and gene-gene interactions contributing to AD risk. Guided by GWAS results, next-generation sequencing and functional studies are under way with the hope of helping us better understand AD pathology and providing new drug targets.
Collapse
Affiliation(s)
- Richard Sherva
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Genetics and Genomics, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Biomedical Genetics L-320, 72 East Concord Street, Boston, MA 02118, USA
| |
Collapse
|
28
|
Elliott DA, Weickert CS, Garner B. Apolipoproteins in the brain: implications for neurological and psychiatric disorders. CLINICAL LIPIDOLOGY 2010; 51:555-573. [PMID: 21423873 PMCID: PMC3058497 DOI: 10.2217/clp.10.37] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The brain is the most lipid-rich organ in the body and, owing to the impermeable nature of the blood-brain barrier, lipid and lipoprotein metabolism within this organ is distinct from the rest of the body. Apolipoproteins play a well-established role in the transport and metabolism of lipids within the CNS; however, evidence is emerging that they also fulfill a number of functions that extend beyond lipid transport and are critical for healthy brain function. The importance of apolipoproteins in brain physiology is highlighted by genetic studies, where apolipoprotein gene polymorphisms have been identified as risk factors for several neurological diseases. Furthermore, the expression of brain apolipoproteins is significantly altered in several brain disorders. The purpose of this article is to provide an up-to-date assessment of the major apolipoproteins found in the brain (ApoE, ApoJ, ApoD and ApoA-I), covering their proposed roles and the factors influencing their level of expression. Particular emphasis is placed on associations with neurological and psychiatric disorders.
Collapse
Affiliation(s)
- David A Elliott
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
| | - Cyndi Shannon Weickert
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
- Schizophrenia Research Institute, Sydney, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Brett Garner
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
- School of Biological Sciences, Faculty of Science, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
29
|
Zoubeidi A, Chi K, Gleave M. Targeting the cytoprotective chaperone, clusterin, for treatment of advanced cancer. Clin Cancer Res 2010; 16:1088-93. [PMID: 20145158 PMCID: PMC2822877 DOI: 10.1158/1078-0432.ccr-09-2917] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many strategies used to kill cancer cells induce stress-responses that activate survival pathways to promote emergence of a treatment resistant phenotype. Secretory clusterin (sCLU) is a stress-activated cytoprotective chaperone up-regulated by many varied anticancer therapies to confer treatment resistance when overexpressed. sCLU levels are increased in several treatment recurrent cancers including castrate resistant prostate cancer, and therefore sCLU has become an attractive target in cancer therapy. sCLU is not druggable with small molecule inhibitors, therefore nucleotide-based strategies to inhibit sCLU at the RNA level are appealing. Preclinical studies have shown that antisense oligonucleotide (ASO) or siRNA knockdown of sCLU have preclinical activity in combination with hormone- and chemotherapy. Phase I and II clinical trial data indicate that the second generation ASO, custirsen (OGX-011), has biologic and clinical activity, suppressing sCLU expression in prostate cancer tissues by more than 90%. A randomized study comparing docetaxel-custirsen to docetaxel alone in men with castrate resistant prostate cancer reported improved survival by 7 months from 16.9 to 23.8 months. Strong preclinical and clinical proof-of-principle data provide rationale for further study of sCLU inhibitors in randomized phase III trials, which are planned to begin in 2010.
Collapse
Affiliation(s)
- Amina Zoubeidi
- Department of Urological Sciences, The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
30
|
Rizzi F, Coletta M, Bettuzzi S. Chapter 2: Clusterin (CLU): From one gene and two transcripts to many proteins. Adv Cancer Res 2010; 104:9-23. [PMID: 19878770 DOI: 10.1016/s0065-230x(09)04002-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Clusterin (CLU) has kept many researchers engaged for a long time since its first discovery and characterization in the attempt to unravel its biological role in mammals. Although there is a general consensus on the fact that CLU is supposed to play important roles in nearly all fundamental biological phenomena and in many human diseases including cancer, after about 10 years of work CLU has been defined as an "enigmatic" protein. This sense of frustration among the researchers is originated by the fact that, despite considerable scientific production concerning CLU, there is still a lack of basic information about the complex regulation of its expression. The CLU gene is a single 9-exon gene expressed at very different levels in almost all major tissues in mammals. The gene produces at least three protein forms with different subcellular localization and diverse biological functions. The molecular mechanism of production of these protein forms remains unclear. The best known is the glycosylated mature form of CLU (sCLU), secreted with very big quantitative differences at different body sites. Hormones and growth factors are the most important regulators of CLU gene expression. Before 2006, it was believed that a unique transcript of about 1.9 kb was originated by transcription of the CLU gene. Now we know that alternative transcriptional initiation, possibly driven by two distinct promoters, may produce at least two distinct CLU mRNA isoforms differing in their unique first exon, named Isoform 1 and Isoform 2. A third transcript, named Isoform 11036, has been recently found as one of the most probable mRNA variants. Approaches like cloning, expression, and functional characterization of the different CLU protein products have generated a critical mass of information teaching us an important lesson about CLU gene expression regulation. Nevertheless, further studies are necessary to better understand the tissue-specific regulation of CLU expression and to identify the specific signals triggering the expression of different/alternative transcript isoforms and protein forms in different cell types at appropriate time.
Collapse
Affiliation(s)
- Federica Rizzi
- Dipartimento di Medicina Sperimentale, Sezione di Biochimica, Biochimica Clinica e Biochimica dell'Esercizio Fisico, Parma, Italy
| | | | | |
Collapse
|
31
|
Nuutinen T, Suuronen T, Kauppinen A, Salminen A. Clusterin: a forgotten player in Alzheimer's disease. ACTA ACUST UNITED AC 2009; 61:89-104. [PMID: 19651157 DOI: 10.1016/j.brainresrev.2009.05.007] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 05/13/2009] [Accepted: 05/14/2009] [Indexed: 11/16/2022]
Abstract
Clusterin, also known as apolipoprotein J, is a versatile chaperone molecule which contains several amphipathic and coiled-coil alpha-helices, typical characteristics of small heat shock proteins. In addition, clusterin has three large intrinsic disordered regions, so-called molten globule domains, which can stabilize stressed protein structures. Twenty years ago, it was demonstrated that the expression of clusterin was clearly increased in Alzheimer's disease (AD). Later it was observed that clusterin can bind amyloid-beta peptides and prevent their fibrillization. Clusterin is also involved in the clearance of amyloid-beta peptides and fibrils by binding to megalin receptors and enhancing their endocytosis within glial cells. Clusterin is a complement inhibitor and can suppress complement activation observed in AD. Clusterin is also present in lipoprotein particles and regulates cholesterol and lipid metabolism of brain which is disturbed in AD. Clusterin is a stress-induced chaperone which is normally secreted but in conditions of cellular stress, it can be transported to cytoplasm where it can bind to Bax protein and inhibit neuronal apoptosis. Clusterin can also bind to Smad2/3 proteins and potentiate the neuroprotective TGFbeta signaling. An alternative splicing can produce a variant isoform of clusterin which can be translocated to nuclei where it induces apoptosis. The role of nuclear clusterin in AD needs to be elucidated. We will review here the extensive literature linking clusterin to AD and examine the recent progress in clusterin research with the respect to AD pathology. Though clusterin can be viewed as a multipotent guardian of brain, it is unable to prevent the progressive neuropathology in chronic AD.
Collapse
Affiliation(s)
- Tapio Nuutinen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | | | | | | |
Collapse
|
32
|
Garcia-Reyero N, Kroll KJ, Liu L, Orlando EF, Watanabe KH, Sepúlveda MS, Villeneuve DL, Perkins EJ, Ankley GT, Denslow ND. Gene expression responses in male fathead minnows exposed to binary mixtures of an estrogen and antiestrogen. BMC Genomics 2009; 10:308. [PMID: 19594897 PMCID: PMC2713996 DOI: 10.1186/1471-2164-10-308] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 07/13/2009] [Indexed: 12/31/2022] Open
Abstract
Background Aquatic organisms are continuously exposed to complex mixtures of chemicals, many of which can interfere with their endocrine system, resulting in impaired reproduction, development or survival, among others. In order to analyze the effects and mechanisms of action of estrogen/anti-estrogen mixtures, we exposed male fathead minnows (Pimephales promelas) for 48 hours via the water to 2, 5, 10, and 50 ng 17α-ethinylestradiol (EE2)/L, 100 ng ZM 189,154/L (a potent antiestrogen known to block activity of estrogen receptors) or mixtures of 5 or 50 ng EE2/L with 100 ng ZM 189,154/L. We analyzed gene expression changes in the gonad, as well as hormone and vitellogenin plasma levels. Results Steroidogenesis was down-regulated by EE2 as reflected by the reduced plasma levels of testosterone in the exposed fish and down-regulation of genes in the steroidogenic pathway. Microarray analysis of testis of fathead minnows treated with 5 ng EE2/L or with the mixture of 5 ng EE2/L and 100 ng ZM 189,154/L indicated that some of the genes whose expression was changed by EE2 were blocked by ZM 189,154, while others were either not blocked or enhanced by the mixture, generating two distinct expression patterns. Gene ontology and pathway analysis programs were used to determine categories of genes for each expression pattern. Conclusion Our results suggest that response to estrogens occurs via multiple mechanisms, including canonical binding to soluble estrogen receptors, membrane estrogen receptors, and other mechanisms that are not blocked by pure antiestrogens.
Collapse
Affiliation(s)
- Natàlia Garcia-Reyero
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Möller FJ, Zierau O, Hertrampf T, Bliedtner A, Diel P, Vollmer G. Long-term effects of dietary isoflavones on uterine gene expression profiles. J Steroid Biochem Mol Biol 2009; 113:296-303. [PMID: 19429436 DOI: 10.1016/j.jsbmb.2009.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/22/2009] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
Isoflavones (ISOs) are bioactive food ingredients of the traditional East Asian diet and currently discussed as alternatives to classical hormone replacement therapies and for reducing the prevalence of hormone-dependent cancers. Although there are many studies on ISOs, not much is known about their long-term effects. Therefore, we performed an animal experiment analyzing the effects of three different diets: a phytoestrogen-free diet, a diet supplemented with genistein (700 microg/g diet) and an ISO-high diet (232 microg daidzein and 240 microg genistein/g) at two distinct time points, juvenile (21 days) and adult (97 days). Exposure started prior to mating of the parents and throughout the life of the offspring. We observed a stronger increase of uterine wet weights in juvenile offspring with genistein exposure (1018+/-350 mg/kg BW) than with ISO-high diet (497+/-133 mg/kg BW). Whereas the expression of proliferation related genes (PCNA; Ki67; IGF-1; IGF-1R), analyzed by real-time-qPCR and Western blot, were significantly down-regulated in juvenile animals exposed to genistein. Additionally, genistein exposure led to estrogenic responses, observed upon increase of complement C3 and decrease of estrogen receptors gene expressions, while the exposure to ISO-high diet did not show these effects. In conclusion, both the time point on which phytoestrogen exposure starts together with the composition of the ingested phytoestrogen containing diet are of great importance for the biological response of the offspring.
Collapse
Affiliation(s)
- Frank Josef Möller
- Institute for Zoology, Molecular Cell Physiology and Endocrinology, Technical University Dresden, Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
Differential expression of clusterin according to histological type of endometrial carcinoma. Gynecol Oncol 2008; 110:222-9. [DOI: 10.1016/j.ygyno.2008.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Revised: 04/05/2008] [Accepted: 04/10/2008] [Indexed: 11/21/2022]
|
35
|
Neuroprotection: VEGF, IL-6, and clusterin: the dark side of the moon. PROGRESS IN BRAIN RESEARCH 2008; 173:555-73. [PMID: 18929134 DOI: 10.1016/s0079-6123(08)01138-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Growth factors and their respective receptors are key regulators in development and homeostasis of the nervous system, and changes in the function, expression, or downstream signaling of growth factors are involved in many neuropathological disorders. Recently, research has yielded a rich harvest of information about molecules and gene, and currently the assumption "a gene-a protein", where each gene encodes the structure of a single protein, is becoming a paradox. In the past years, the discovery of synergic or antagonistic proteins deriving from the same gene is a novelty upsetting. In some way, the conventional function of proteins involved in DNA repair, cell death/growth induction, vascularization, and metabolism is inhibited or shifted toward other pathways by soluble mediators that orchestrate such change depending on the microenvironment conditions. In this chapter, we focus on the antithetic properties that proteins could exert, depending on the microenvironment that orchestrates the complex networks among proteins and their respective partners.
Collapse
|
36
|
Andersen CL, Schepeler T, Thorsen K, Birkenkamp-Demtröder K, Mansilla F, Aaltonen LA, Laurberg S, Ørntoft TF. Clusterin Expression in Normal Mucosa and Colorectal Cancer. Mol Cell Proteomics 2007; 6:1039-48. [PMID: 17322305 DOI: 10.1074/mcp.m600261-mcp200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The gene Clusterin is a target for cancer therapy in clinical trials. The indication for intervention is up-regulated Clusterin expression. Clusterin has been reported to be deregulated in multiple cancer types, including colorectal cancer (CRC). However, for CRC the studies have disagreed on whether Clusterin is up- or down-regulated by neoplastic cells. In the present study we sought to clarify the expression and distribution of Clusterin mRNAs and proteins in normal and neoplastic colorectal tissue through laser microdissection, variant-specific real time RT-PCR, immunohistochemistry, immunofluorescence, Western blotting, and array-based transcriptional profiling. At the transcript level we demonstrated the expression of two novel Clusterin transcripts in addition to the known transcript, and at the protein level we demonstrated two Clusterin isoforms. Our analysis of normal epithelial cells revealed that among these, Clusterin was only expressed by rare neuroendocrine subtype. Furthermore our analysis showed that in the normal mucosa the majority of the observed Clusterin protein originated from the stromal compartment. In tumors we found that Clusterin was de novo synthesized by non-neuroendocrine cancer cells in approximately 25% of cases. Moreover we found that the overall Clusterin level in tumors often appeared to be lower than in normal mucosa due to the stromal compartment often being suppressed in tumors. Although Clusterin in normal neuroendocrine cells showed a basal localization, the localization in cancer cells was often apical and in some cases associated with apical secretion. Collectively our results indicate that Clusterin expression is very complex. We conclude that Clusterin expression is associated with neuroendocrine differentiation in normal epithelia and that the Clusterin observed in neoplastic cells is de novo synthesized. The cases with de novo synthesized Clusterin define a distinct subgroup of CRC that may be of clinical importance as anti-Clusterin therapeutics are now in clinical trials.
Collapse
Affiliation(s)
- Claus Lindbjerg Andersen
- Molecular Diagnostic Laboratory, Department of Clinical Biochemistry, Aarhus University Hospital, DK8200 Aarhus N, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Savković V, Gantzer H, Reiser U, Selig L, Gaiser S, Sack U, Klöppel G, Mössner J, Keim V, Horn F, Bödeker H. Clusterin is protective in pancreatitis through anti-apoptotic and anti-inflammatory properties. Biochem Biophys Res Commun 2007; 356:431-7. [PMID: 17359935 DOI: 10.1016/j.bbrc.2007.02.148] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2007] [Accepted: 02/27/2007] [Indexed: 11/26/2022]
Abstract
Clusterin is overexpressed in pancreas during the acute phase of pancreatitis. We intended to clarify the role of clusterin expression in stressed exocrine pancreas. We performed in vitro experiments in transfected AR4-2J cells with modified expression levels of clusterin and in vivo studies in clusterin-deficient mice. AR4-2J cells were exposed to agents mimicking cell-stress during pancreatitis (cerulein, hydrogen peroxide, staurosporine or lysophosphatidylcholine). Clusterin-overexpressing AR4-2J cells showed higher viability after cell stress and accordingly reduced rates of apoptosis and lessened caspase-3 activation. Blockage of endogenous clusterin expression reduced viability and enhanced apoptosis. Presence of clusterin reduced NF-kappaB activation and expression of the NF-kappaB target genes TNF-alpha and MOB-1 under cell stress. Clusterin-deficient mice showed a more severe course of acute experimental pancreatitis with enhanced rates of apoptosis and inflammatory cell infiltration. We concluded that clusterin was protective during inflammation of exocrine pancreas because of its anti-apoptotic and anti-inflammatory functions.
Collapse
Affiliation(s)
- Vuk Savković
- Medizinische Klinik und Poliklinik 2, Universitätsklinikum Leipzig AöR, Ph.-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cochrane DR, Wang Z, Muramaki M, Gleave ME, Nelson CC. Differential regulation of clusterin and its isoforms by androgens in prostate cells. J Biol Chem 2006; 282:2278-87. [PMID: 17148459 DOI: 10.1074/jbc.m608162200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Clusterin mRNA levels were shown to increase dramatically in rat ventral prostate following castration, and clusterin was therefore originally thought to be repressed by androgens. It was later discovered that the increased clusterin levels are most likely due to castration-induced apoptosis of the prostatic epithelium rather than direct action of the androgen receptor (AR). In the studies presented here, LNCaP cells in culture and rat prostate organ culture were treated with androgens. Clusterin mRNA and protein are shown to increase with androgen treatment in a time- and dose-dependent manner. This induction of clusterin requires AR and can be inhibited by casodex, an AR antagonist. We have found that the first intron of the clusterin gene contains putative androgen response elements. The intronic region is shown to be bound by AR in chromatin immunoprecipitation assays and is transactivated by AR in reporter assays. Two isoforms of clusterin result from alternate transcriptional start sites. Both isoforms are cytoprotective; however, Isoform 1 has the capacity to produce a splice variant that is apoptotic. Real time PCR was used to determine the response of the two isoforms to androgens. Intriguingly, these results illustrated that Isoform 2 was up-regulated, whereas Isoform 1 was down-regulated by androgens. Isoform 2 was also increased as the LNCaP xenograft tumor progressed to androgen-independence, whereas Isoform 1 was unaltered. This androgen regulation of clusterin may underline the cytoprotective role of androgens in normal prostate physiology as well as play an antiapoptotic role in prostate cancer progression.
Collapse
Affiliation(s)
- Dawn R Cochrane
- Department of Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | |
Collapse
|
39
|
Strocchi P, Smith MA, Perry G, Tamagno E, Danni O, Pession A, Gaiba A, Dozza B. Clusterin up-regulation following sub-lethal oxidative stress and lipid peroxidation in human neuroblastoma cells. Neurobiol Aging 2006; 27:1588-94. [PMID: 16464517 DOI: 10.1016/j.neurobiolaging.2005.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2005] [Revised: 09/02/2005] [Accepted: 09/20/2005] [Indexed: 10/25/2022]
Abstract
Clusterin/apolipoprotein J is a multifunctional protein up-regulated during various pathophysiological states. Since oxidative stress plays an important role in brain aging, and in many neurodegenerative disorders, to further understand the mechanistic underpinnings of clusterin expression, in this study, we examined clusterin expression in human neuroblastoma cells under conditions of increased production of reactive oxygen species and lipid peroxidation. Specifically, we analyzed clusterin mRNA and protein levels in human neuroblastoma IMR-32 and SH-SY5Y cells following exposure to sub-lethal amounts of iron-ascorbate to induce an increase in reactive oxygen species generation and lipid peroxidation. Under such conditions, we observed a time-dependent up-regulation of clusterin protein and mRNA levels, detected by immunoblot analysis and RT-PCR, respectively. Given the known roles of clusterin, the results of the present study support the notion that an increase in clusterin expression may be a physiological defence mounted to reduce cell damage and maintain cell viability during periods of increased oxidative stress.
Collapse
Affiliation(s)
- Paola Strocchi
- Department of Pharmacology, University of Bologna, Bologna 40126, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Seubert JM, Xu F, Graves JP, Collins JB, Sieber SO, Paules RS, Kroetz DL, Zeldin DC. Differential renal gene expression in prehypertensive and hypertensive spontaneously hypertensive rats. Am J Physiol Renal Physiol 2005; 289:F552-61. [PMID: 15798089 DOI: 10.1152/ajprenal.00354.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Development of hypertension stems from both environmental and genetic factors wherein the kidney plays a central role. Spontaneously hypertensive rats (SHR) and the nonhypertensive Wistar-Kyoto (WKY) controls are widely used as a model for studying hypertension. The present study examined the renal gene expression profiles between SHR and WKY at a prehypertensive stage (3 wk of age) and hypertensive stage (9 wk of age). Additionally, age-related changes in gene expression patterns were examined from 3 to 9 wk in both WKY and SHR. Five to six individual kidney samples of the same experimental group were pooled together, and quadruplicate hybridizations were performed using the National Institute of Environmental Health Sciences Rat version 2.0 Chip, which contains ∼6,700 genes. Twenty two genes were found to be differentially expressed between SHR and WKY at 3 wk of age, and 104 genes were differentially expressed at 9 wk of age. Soluble epoxide hydrolase ( Ephx2) was found to be significantly upregulated in SHR at both time points and was the predominant outlier. Conversely, elastase 1 ( Ela1) was found to be the predominant gene downregulated in SHR at both time points. Analysis of profiles at 3 vs. 9 wk of age identified 508 differentially expressed genes in WKY rats. In contrast, only 211 genes were found to be differentially expressed during this time period in SHR. The altered gene expression patterns observed in the age-related analysis suggested significant differences in the vascular extracellular matrix system between SHR and WKY kidney. Together, our data highlight the complexity of hypertension and the numerous genes involved in and affected by this condition.
Collapse
Affiliation(s)
- J M Seubert
- Division of Intramural Research, National Center for Toxicogenomics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sundar SV, Li YY, Rollwagen FM, Maheshwari RK. Hemorrhagic shock induces differential gene expression and apoptosis in mouse liver. Biochem Biophys Res Commun 2005; 332:688-96. [PMID: 15907801 DOI: 10.1016/j.bbrc.2005.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 05/05/2005] [Indexed: 01/07/2023]
Abstract
Comprehensive knowledge of the gene expression changes induced by hemorrhage in vital organs will greatly improve prognosis and therapy. Therefore, we used a mouse model of non-resuscitated hemorrhagic shock to study the pattern of stress-induced genes in liver at 1, 4, and 24 h following surgery. Hepatic injury was confirmed by assessment of liver injury markers and apoptotic cell death. We found that a variety of stress-regulated genes were differentially expressed, including seven genes that have not been reported previously as being regulated by hemorrhagic shock: ATF-2, alphaB-crystallin, GADD45, GADD45beta, Mdm2, p21Waf1, and TRPM-2. The changes in mRNA levels of the transcription factors AP-1, Egr-1, HSF-1, and NF-kappaB were transient but protein expression was noticeable at later time points. Our findings show that oxidative stress causes immediate upregulation of genes involved in a variety of cellular defense pathways. Complex interactions among them might determine the ultimate fate of the cell.
Collapse
Affiliation(s)
- Shirin V Sundar
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
42
|
Gleave M, Miyake H. Use of antisense oligonucleotides targeting the cytoprotective gene, clusterin, to enhance androgen- and chemo-sensitivity in prostate cancer. World J Urol 2005; 23:38-46. [PMID: 15770517 DOI: 10.1007/s00345-004-0474-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Accepted: 11/01/2004] [Indexed: 01/13/2023] Open
Abstract
The discovery and targeting of genes mediating androgen-independence may lead to the development of novel therapies that delay progression of hormone refractory prostate cancer (HRPC). Clusterin is a stress-associated cell survival gene that increases after androgen ablation. Here, we review clusterin's functional role in apoptosis and the use of antisense oligonucleotides (ASOs) against clusterin to enhance apoptosis in prostate cancer models. Immunostaining of tissue microarrays constructed from untreated and post-hormone treated radical prostatectomy specimens confirm that clusterin is highly expressed in virtually all HRPC cells, 80% of prostate cancer cells after neoadjuvant hormone therapy, but is low or absent (<20%) in untreated specimens. Overexpression of clusterin in LNCaP cells confers resistance to both androgen ablation and chemotherapy. Clusterin ASOs reduced clusterin levels in a dose-dependent and sequence-specific manner. Adjuvant treatment with murine clusterin ASOs after castration of mice bearing Shionogi tumors decreased clusterin levels, accelerated apoptotic tumor regression, and significantly delayed the recurrence of androgen-independent tumors. A human clusterin ASO targeting the translation initiation site and incorporating MOE-gapmer backbone (OGX-011) synergistically enhanced the cytotoxic effects of paclitaxel in human xenografts of prostate, renal cell, bladder, and lung cancer. Clusterin, is an anti-apoptosis protein upregulated in an adaptive cell survival manner by androgen ablation and chemotherapy that confers resistance to various cell death triggers. Suppression of clusterin levels using ASOs enhances cell death following treatment with androgen ablation, radiation, and chemotherapy.
Collapse
Affiliation(s)
- Martin Gleave
- Division of Urology, University of British Columbia, D-9, 2733 Heather Street, Vancouver, British Columbia , V5Z 3J5, Canada.
| | | |
Collapse
|
43
|
Guth SE, Böhm S, Mussler BH, Eisenbrand G. Sensitivein vitro test systems to determine androgenic/antiandrogenic activity. Mol Nutr Food Res 2004; 48:282-91. [PMID: 15497179 DOI: 10.1002/mnfr.200400023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We report on the establishment of one transgenic and two endogenous reporter gene assays to determine androgenic/antiandrogenic activity. A transient transactivation assay was developed in COS-7 African green monkey kidney cells. Three plasmids were co-transfected by electroporation: the human androgen receptor expression vector pSG5AR, the reporter gene vector pMamneoLuc, expressing luciferase under the control of the mouse mammary tumor virus (MMTV) promotor containing 4 hormone responsive elements (HREs), and the control plasmid pSVbeta. Transcriptional activation was measured by luciferase-mediated chemoluminescence. In T47D human breast cancer cells two endogenous reporter gene systems were established: one based on the androgen-induced expression of prostate-specific antigen (PSA), the other based on the androgen-repressed expression of testosterone repressed message 2 (TRPM-2). PSA protein was measured by enzyme-linked immunosorbent assay (ELISA), TRPM-2 m-RNA by reverse transcriptase polymerase chain reaction (RT-PCR). All three test systems were validated using the physiological androgen dihydrotestosterone (DHT) as agonist and the established antiandrogens hydroxyflutamide and p,p'-dichlorodiphenylethene (p,p'-DDE) as antagonists. The PSA assay was the most sensitive test system with an EC50 of 0.7 nM for DHT-induced response. The transient transactivation assay in COS-7 cells was less sensitive with an EC50 of 9.7 nM DHT. In the PSA assay hydroxyflutamide was a more potent antagonist (IC30 = 0.02 microM) than p,p'-DDE (IC30 = 0.9 microM). In the transient transactivation assay in COS-7 cells, both compounds elicited about 30% of the agonistic response induced by 100 nM DHT, thus showing partial agonistic properties. In summary, three highly sensitive and complementary in vitro test systems, together achieving enhanced specificity for detection and assessment of androgenic/antiandrogenic activity have been established and validated.
Collapse
Affiliation(s)
- Sabine E Guth
- Department of Chemistry, Division of Food Chemistry and Environmental Toxicology, University of Kaiserslautern, Kaiserslautem, Germany
| | | | | | | |
Collapse
|
44
|
Bayon Y, Ortiz MA, Lopez-Hernandez FJ, Howe PH, Piedrafita FJ. The Retinoid Antagonist MX781 Induces Clusterin Expression in Prostate Cancer Cells via Heat Shock Factor-1 and Activator Protein-1 Transcription Factors. Cancer Res 2004; 64:5905-12. [PMID: 15313936 DOI: 10.1158/0008-5472.can-03-3657] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Retinoids mediate numerous biological responses through the transcriptional activation of nuclear retinoid receptors. Due to their antiproliferative activity, retinoids have shown promise as anticancer agents. Synthetic analogs have been described that selectively activate one subset of the retinoid receptors or inhibit their transcriptional activity. Some of these compounds exhibit strong anticancer activity, which is associated with their ability to induce apoptosis. Here we describe that the retinoid antagonist MX781 causes a substantial increase of clusterin mRNA and protein levels in prostate carcinoma cells. In contrast, retinoic acid and other synthetic agonists and antagonists show no effect on clusterin mRNA/protein levels. Induction of clusterin mRNA is associated with transcriptional activation of the clusterin promoter, which requires the proximal -218-bp region containing binding sites for heat shock factor (HSF)-1, activator protein (AP)-2, and AP-1 transcription factors. MX781 slightly induces AP-1 DNA binding activity, and mutation of the AP-1 site differentially affects the activation of the clusterin promoter in a cell type-specific manner. In contrast, a robust increase of HSF-1 DNA binding activity is observed in all cancer cell lines examined, and mutation of the heat shock element site in the clusterin promoter completely abolishes MX781-induced transcriptional activation in PC3 and DU145 cells. Other agonist retinoid-related molecules also induce AP-1 activity, but not HSF-1, and elicit no effect on clusterin expression levels. These data point to HSF-1 as an important factor regulating clusterin expression in response to MX781, although AP-1 activity may also participate in a cell type-specific manner.
Collapse
Affiliation(s)
- Yolanda Bayon
- Sidney Kimmel Cancer Center, San Diego, California 92121, USA.
| | | | | | | | | |
Collapse
|
45
|
Pucci S, Bonanno E, Pichiorri F, Angeloni C, Spagnoli LG. Modulation of different clusterin isoforms in human colon tumorigenesis. Oncogene 2004; 23:2298-304. [PMID: 14755245 DOI: 10.1038/sj.onc.1207404] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Clusterin is a ubiquitous secretory heterodimeric disulfide-linked glycoprotein, which is implicated in several physiological processes, including immune regulation, cell adhesion and morphological transformation, lipid transportation, tissue remodelling, membrane recycling and cell-cell interactions. A large number of studies have focused their interest on clusterin gene products as mediators of cell cycle progression and cell death induction, although data on the different isoforms and their role in the different cell processes are still obscure. Recently, an increased clusterin expression in breast cancer has been reported. In order to elucidate the role of clusterin in tumor progression and whether one of its isoforms is preferentially expressed in tumorigenesis, we examined its presence throughout the different steps of human colon carcinoma, one of the best-characterized models of human tumor progression. The immunohistochemical observation of 30 bioptic and surgical colon specimens demonstrated a cell compartment clusterin translocation from the nucleus to the cytoplasm directly related to tumor progression. In fact, a nuclear localization found in healthy colonic mucosa is consistent with the involvement of the proapoptotic nuclear form in the regulation of cell cycle progression and in cell death induction. The progression towards high-grade and metastatic carcinoma leads to cytoplasmic clusterin distribution. Protein extracts from freshly isolated cells of the same patients confirm in high-grade carcinomas with metastatic nodes the complete loss of the proapoptotic nuclear form and a cytoplasmic overexpression of the highly glycosylated form. Data obtained from in vitro experiments confirm that this form is released in the extracellular space and corresponded to the fully glycosylated one. These data suggest that the controversial data on clusterin function in tumors may be related to the pattern shift of its isoform production. As the secreted form of clusterin is correlated to cell matrix formation, cell membrane remodeling and cell-cell adhesion, the overexpression of this form in highly aggressive tumors and metastatic nodes could be a potential new prognostic and predictive marker for colon carcinoma aggressiveness.
Collapse
Affiliation(s)
- Sabina Pucci
- Department of Biopathology, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy.
| | | | | | | | | |
Collapse
|
46
|
Abstract
The primary hurdle to improved survival of advanced prostate cancer is our failure to prevent or treat the tumor's progression to its lethal and untreatable stage of androgen independence. Novel treatment modalities designed to prevent androgen-independent progression including prostate cancer metastasis are required. Accelerated identification and characterization of cancer-relevant molecular targets has sparked considerable interest in the development of new generations of anticancer agents that specifically inhibit a progression-relevant target. Antisense oligonucleotides, short synthetic stretches of chemically modified DNA capable of specifically hybridizing to the mRNA of a chosen cancer-relevant target gene, promise to show enhanced specificity for malignant cells with a favorable side-effect profile due to well-defined and tailored modes of action. Although not all of the challenges have been met to date, emerging clinical evidence supports the premise that antisense oligonucleotides stand a realistic chance of emerging as major partners of rationally designed anticancer regimens. The rationale and status of antisense targeting of the treatment resistance factor clusterin and of insulin-like growth factor binding protein (IGFBP) 2 and 5 are discussed.
Collapse
Affiliation(s)
- Martin Gleave
- The Prostate Centre at Vancouver General Hospital, Division of Urology, University of British Columbia, Vancouver, BC, Canada, V6H 3Z3.
| | | |
Collapse
|
47
|
Abstract
Coronary heart disease is one of the major causes of death in developed countries. The hypothesis that peroxidation of low density lipoproteins (LDL) may be the initial step of the atherosclerotic process has promoted numerous studies aimed at investigating the mechanisms by which the body protects itself from such oxidative phenomena. Among these mechanisms we find the paraoxanase (PON) enzyme, which is quite thriving the last decades. This enzyme is principally associated with high density lipoproteins (HDL) but it also seems to help LDL to recover their antioxidant status. This paper reviews different aspects concerning the mechanisms implicated in the induction and activity of this enzyme, as well as its production, attachment to HDL, and modifications of its activity due to external factors. The use of genetic techniques, the study of the polimorphisms of the PON enzyme and the possibility of increasing paraoxonase activity by means of pharmacotherapy and/or dietary therapy open new perspectives with regard to coronary heart disease treatment and prevention.
Collapse
Affiliation(s)
- Amaia Canales
- Departamento de Nutrición. Facultad de Farmacia. Universidad Complutense de Madrid. Madrid. España
| | | |
Collapse
|
48
|
Zellweger T, Kiyama S, Chi K, Miyake H, Adomat H, Skov K, Gleave ME. Overexpression of the cytoprotective protein clusterin decreases radiosensitivity in the human LNCaP prostate tumour model. BJU Int 2003; 92:463-9. [PMID: 12930442 DOI: 10.1046/j.1464-410x.2003.04349.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the effect of clusterin overexpression on radiation-induced tumour growth rates and apoptosis in human prostate LNCaP cells, as prostate cancer cells are relatively resistant to radiation-induced apoptosis and local recurrences are common, but overexpression of the anti-apoptotic protein clusterin can accelerate progression to androgen-independence and to confer a chemoresistant phenotype in various prostate cancer models. MATERIALS AND METHODS Western blot analysis and immunohistochemistry were used to compare clusterin expression levels in parental (P) and clusterin-transfected (T) LNCaP cells in vitro and in vivo. The effects of radiation on clusterin-expression in both parental LNCaP/P and clusterin-transfected LNCaP/T tumours were analysed by Northern blot analysis. The cellular response to radiation was determined up to 3 weeks after irradiation using tetrazolium and re-growth assays, and cell-cycle analysis by flow cytometry. RESULTS Clusterin mRNA expression increased from undetectable to low levels in LNCaP/P tumours after radiation and more than three-fold in LNCaP/T tumours. Clusterin overexpression decreased the radiosensitivity in a time-dependent manner, reducing the extent of growth arrest and apoptosis by up to 54%. Re-growth assays showed that the improved survival rates of LNCaP/T cells after radiation did not change after 3 days, remaining constant over 3 weeks. CONCLUSIONS These results identify clusterin as a promoter of cell survival that may help mediate resistance to radiation-induced apoptosis. Furthermore, clusterin overexpression seems to provide an extended protection against radiation-induced cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- T Zellweger
- The Prostate Centre, Vancouver General Hospital, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Chen X, Halberg RB, Ehrhardt WM, Torrealba J, Dove WF. Clusterin as a biomarker in murine and human intestinal neoplasia. Proc Natl Acad Sci U S A 2003; 100:9530-5. [PMID: 12886021 PMCID: PMC170952 DOI: 10.1073/pnas.1233633100] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Early detection of colorectal cancer is critical for the management of this disease. Biomarkers for early detection of several cancers have been developed and applied clinically in recent years. We have sought to discover candidate biomarkers without the restricted choice of markers placed on microarrays, and without the biological complications of genetic and environmental heterogeneity. We have compared by cDNA subtraction two genetically matched sets of mice, one developing multiple intestinal neoplasia (C57BL/6J-ApcMin) and the other tumor-free (C57BL/6J). One prominent candidate biomarker, clusterin, was then subjected to a series of validation steps. In situ hybridization and immunohistochemistry were used to analyze clusterin expression at a cellular level on a series of murine intestinal and human colonic neoplasms. Elevated clusterin expression was characterized within certain regions of murine and human tumors regardless of tumor stage, location, or mode of initiation. The cells showing high clusterin levels generally lacked differentiation markers and adenomatous polyposis coli antigen. Tumor cells undergoing apoptosis expressed low levels of clusterin. Its specific expression patterns and correlation with cellular events during tumorigenesis make it a useful diagnostic tool in the mouse and a potential contributor to the set of biomarkers for early detection of human colon cancer.
Collapse
Affiliation(s)
- Xiaodi Chen
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
50
|
O'Sullivan J, Whyte L, Drake J, Tenniswood M. Alterations in the post-translational modification and intracellular trafficking of clusterin in MCF-7 cells during apoptosis. Cell Death Differ 2003; 10:914-27. [PMID: 12867999 DOI: 10.1038/sj.cdd.4401254] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Clusterin is a heterodimeric, disulfide-linked 70-80 kDa glycoprotein that is induced during regression of most, if not all, hormone-dependent epithelial tissues. These studies describe the biogenesis and intracellular trafficking of clusterin in MCF-7 cells before and after the initiation of apoptosis with antiestrogens and TNF alpha. Under physiological conditions, clusterin is modified in the endoplasmic reticulum (ER), and proteolytically cleaved in the Golgi to generate discrete alpha and beta chains prior to secretion. Treatment with TNFalpha or the antiestrogen, ICI 182,780, induces apoptosis in MCF-7 cells and leads to substantial changes in the activity of Golgi-resident enzymes, significantly altering the biogenesis of clusterin. This leads to the appearance of a 50-53 kDa uncleaved, nonglycosylated, disulfide-linked isoform of clusterin that accumulates in the nucleus. While clusterin contains a cryptic SV-40-like nuclear localization signal, mutation of this sequence does not affect the nuclear accumulation of the disulfide-linked nuclear isoform. Confocal microscopy demonstrates that the nuclear accumulation of clusterin is coincident with DNA fragmentation. These data suggest that, at least in secretory epithelial cells, retrograde transport from the Golgi to the ER of a nonglycosylated, uncleaved isoform and the subsequent translocation of clusterin to the nucleus occur in dying cells.
Collapse
Affiliation(s)
- J O'Sullivan
- Department of Biological Sciences, University of Notre Dame, Notre Dame IN 46556, USA
| | | | | | | |
Collapse
|