1
|
Zoccali C, Capasso G. Genetic biomarkers of cognitive impairment and dementia of potential interest in CKD patients. J Nephrol 2024; 37:2473-2479. [PMID: 38970746 DOI: 10.1007/s40620-024-02006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/15/2024] [Indexed: 07/08/2024]
Abstract
This review discusses genetic variants associated with cognitive dysfunction in chronic kidney disease (CKD) patients, emphasising the limited research in this area. Four studies have explored genetic markers of cognitive dysfunction in CKD, with findings suggesting shared genetic biomarkers between Alzheimer's Disease and CKD.Because of the limited specific research on genetic markers of cognitive dysfunction and dementia in CKD, we extracted data from the current literature studies on genetic markers in the general population that may be relevant to the CKD population. These markers include Apolipoprotein E (APOE), Complement Receptor 1 (CR1), Clusterin (CLU), Sortilin-related receptor 1 (SORL1), Catechol-O-methyltransferase (COMT), and Brain-derived neurotrophic factor (BDNF), all of which are known to be associated with cognitive dysfunction and dementia in other populations. These genes play various roles in lipid metabolism, inflammation, Aβ clearance, and neuronal function, making them potential candidates for studying cognitive decline in CKD patients.CKD-specific research is needed to understand the role of these genetic markers in CKD-related cognitive dysfunction. Investigating how these genes influence cognitive decline in CKD patients could provide valuable insights into early detection, targeted interventions, and personalised treatment strategies. Overall, genetic studies to enhance our understanding and management of cognitive dysfunction in CKD represent a clinical research priority in this population.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, USA.
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy.
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy.
| | - Giovambattista Capasso
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
2
|
Palihati N, Tang Y, Yin Y, Yu D, Liu G, Quan Z, Ni J, Yan Y, Qing H. Clusterin is a Potential Therapeutic Target in Alzheimer's Disease. Mol Neurobiol 2024; 61:3836-3850. [PMID: 38017342 DOI: 10.1007/s12035-023-03801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
In recent years, Clusterin, a glycosylated protein with multiple biological functions, has attracted extensive research attention. It is closely associated with the physiological and pathological states within the organism. Particularly in Alzheimer's disease (AD) research, Clusterin plays a significant role in the disease's occurrence and progression. Numerous studies have demonstrated a close association between Clusterin and AD. Firstly, the expression level of Clusterin in the brain tissue of AD patients is closely related to pathological progression. Secondly, Clusterin is involved in the deposition and formation of β-amyloid, which is a crucial process in AD development. Furthermore, Clusterin may affect the pathogenesis of AD through mechanisms such as regulating inflammation, controlling cell apoptosis, and clearing pathological proteins. Therefore, further research on the relationship between Clusterin and AD will contribute to a deeper understanding of the etiology of this neurodegenerative disease and provide a theoretical basis for developing early diagnostic and therapeutic strategies for AD. This also makes Clusterin one of the research focuses as a potential biomarker for AD diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Nazhakaiti Palihati
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Ding Yu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China.
| |
Collapse
|
3
|
Martín-García D, García-Aranda M, Redondo M. Therapeutic Potential of Clusterin Inhibition in Human Cancer. Cells 2024; 13:665. [PMID: 38667280 PMCID: PMC11049052 DOI: 10.3390/cells13080665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Clusterin (CLU) protein is involved in various pathophysiological processes including carcinogenesis and tumor progression. In recent years, the role of the secretory isoform has been demonstrated in tumor cells, where it inhibits apoptosis and favors the acquisition of resistance to conventional treatments used to treat cancer. To determine the possible therapeutic potential of inhibiting this protein, numerous studies have been carried out in this field. In this article, we present the existing knowledge to date on the inhibition of this protein in different types of cancer and analyze the importance it could have in the development of new therapies targeted against this disease.
Collapse
Affiliation(s)
- Desirée Martín-García
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain;
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Málaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Málaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| | - Marilina García-Aranda
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Málaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Málaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| | - Maximino Redondo
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain;
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Málaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Málaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| |
Collapse
|
4
|
Duan S, Qin N, Pi J, Sun P, Gao Y, Liu L, Li Z, Li Y, Shi L, Gao Q, Qiu Y, Tang S, Wang CH, Chen TY, Wang ST, Young KC, Sun HY. Antagonizing apolipoprotein J chaperone promotes proteasomal degradation of mTOR and relieves hepatic lipid deposition. Hepatology 2023; 78:1182-1199. [PMID: 37013405 DOI: 10.1097/hep.0000000000000185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/26/2022] [Indexed: 04/05/2023]
Abstract
BACKGROUND AND AIMS Overnutrition-induced activation of mammalian target of rapamycin (mTOR) dysregulates intracellular lipid metabolism and contributes to hepatic lipid deposition. Apolipoprotein J (ApoJ) is a molecular chaperone and participates in pathogen-induced and nutrient-induced lipid accumulation. This study investigates the mechanism of ApoJ-regulated ubiquitin-proteasomal degradation of mTOR, and a proof-of-concept ApoJ antagonist peptide is proposed to relieve hepatic steatosis. APPROACH AND RESULTS By using omics approaches, upregulation of ApoJ was found in high-fat medium-fed hepatocytes and livers of patients with NAFLD. Hepatic ApoJ level associated with the levels of mTOR and protein markers of autophagy and correlated positively with lipid contents in the liver of mice. Functionally, nonsecreted intracellular ApoJ bound to mTOR kinase domain and prevented mTOR ubiquitination by interfering FBW7 ubiquitin ligase interaction through its R324 residue. In vitro and in vivo gain-of-function or loss-of-function analysis further demonstrated that targeting ApoJ promotes proteasomal degradation of mTOR, restores lipophagy and lysosomal activity, thus prevents hepatic lipid deposition. Moreover, an antagonist peptide with a dissociation constant (Kd) of 2.54 µM interacted with stress-induced ApoJ and improved hepatic pathology, serum lipid and glucose homeostasis, and insulin sensitivity in mice with NAFLD or type II diabetes mellitus. CONCLUSIONS ApoJ antagonist peptide might be a potential therapeutic against lipid-associated metabolic disorders through restoring mTOR and FBW7 interaction and facilitating ubiquitin-proteasomal degradation of mTOR.
Collapse
Affiliation(s)
- Shuangdi Duan
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Nong Qin
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Jiayi Pi
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Pei Sun
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Yating Gao
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Lamei Liu
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Zenghui Li
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Ya Li
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Liyang Shi
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Qiang Gao
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Ye Qiu
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Songqing Tang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Chun-Hsiang Wang
- Division of Gastroenterology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Tzu-Ying Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sin-Tian Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kung-Chia Young
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Yu Sun
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Téllez T, Martin-García D, Redondo M, García-Aranda M. Clusterin Expression in Colorectal Carcinomas. Int J Mol Sci 2023; 24:14641. [PMID: 37834086 PMCID: PMC10572822 DOI: 10.3390/ijms241914641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Colorectal cancer is the third most diagnosed cancer, behind only breast and lung cancer. In terms of overall mortality, it ranks second due to, among other factors, problems with screening programs, which means that one of the factors that directly impacts survival and treatment success is early detection of the disease. Clusterin (CLU) is a molecular chaperone that has been linked to tumorigenesis, cancer progression and resistance to anticancer treatments, which has made it a promising drug target. However, it is still necessary to continue this line of research and to adjust the situations in which its use is more favorable. The aim of this paper is to review the current genetic knowledge on the role of CLU in tumorigenesis and cancer progression in general, and discuss its possible use as a therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Teresa Téllez
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain; (T.T.); (D.M.-G.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
| | - Desirée Martin-García
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain; (T.T.); (D.M.-G.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| | - Maximino Redondo
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain; (T.T.); (D.M.-G.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| | - Marilina García-Aranda
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain;
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| |
Collapse
|
6
|
Satapathy S, Walker H, Brown J, Gambin Y, Wilson MR. The N-end rule pathway regulates ER stress-induced clusterin release to the cytosol where it directs misfolded proteins for degradation. Cell Rep 2023; 42:113059. [PMID: 37660295 DOI: 10.1016/j.celrep.2023.113059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/14/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Previous work suggests that cell stress induces release of the normally secreted chaperone clusterin (CLU) into the cytosol. We analyzed the localization of CLU in healthy and stressed cells, the mechanism of its cytosolic release, and its interactions with cytosolic misfolded proteins. Key results of this study are the following: (1) full-length CLU is released to the cytosol during stress, (2) the CLU N-terminal D1 residue is recognized by the N-end rule pathway and together with the enzyme ATE1 is essential for cytosolic release, (3) CLU can form stable complexes with cytosolic misfolded proteins and direct them to the proteasome and autophagosomes, and (4) cytosolic CLU protects cells from hypoxic stress and the cytosolic overexpression of an aggregation-prone protein. Collectively, the results suggest that enhanced cytosolic release of CLU is a stress response that can inhibit the toxicity of misfolded proteins and facilitate their targeted degradation via both autophagy and the proteasome.
Collapse
Affiliation(s)
- Sandeep Satapathy
- The Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Holly Walker
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - James Brown
- EMBL Australia Node in Single Molecule Science, and School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, and School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark R Wilson
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
7
|
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
|
8
|
Sen S, Udaya P, Maheshwari JJ, Ramasamy K, Kannan NB, Dharmalingam K. Profiling of idiopathic macular hole vitreous proteome identifies the role of extracellular matrix remodelling, epithelial-mesenchymal transformation and unfolded protein-response pathways. Indian J Ophthalmol 2023; 71:2027-2040. [PMID: 37203077 PMCID: PMC10391365 DOI: 10.4103/ijo.ijo_2068_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023] Open
Abstract
Purpose To analyze and describe the proteome of the vitreous humour in eyes with idiopathic macular holes. Methods We performed mass spectrometry (MS)-based label-free quantitative analysis of the vitreous proteome of idiopathic macular hole (IMH) and control donor vitreous. Comparative quantification was performed using SCAFFOLD software which calculated fold changes of differential expression. Bioinformatics analysis was performed using DAVID and STRING software. Results A total of 448 proteins were identified by LC-MS/MS in IMH and cadaveric eye vitreous samples, of which 199 proteins were common. IMH samples had 189 proteins that were unique and 60 proteins were present only in the control cadaveric vitreous. We found upregulation of several extracellular matrix (ECM) and cytoskeletal proteins, namely collagen alpha-1 (XVIII) chain, N-cadherin, EFEMP1/fibulin-3, basement membrane-specific heparan sulfate proteoglycan core protein, and target of Nesh-3. Several cytoskeleton proteins, namely tubulin, actin, and fibronectin levels, were significantly lower in IMH vitreous, probably reflecting increased ECM degradation. IMH vitreous also had a downregulation of unfolded protein response-mediated-mediated apoptosis proteins, possibly related to a state of increased cell survival and proliferation, along with a remodelling and aberrant production of ECM contents. Conclusion The pathogenesis of macular holes may involve ECM remodelling, epithelial-mesenchymal transformation, downregulation of apoptosis, protein folding defects, and complement pathway. The vitreo-retinal milieu in macular holes contain molecules related to both ECM degradation and inhibition of the same, thereby maintaining a homeostasis.
Collapse
Affiliation(s)
- Sagnik Sen
- Department of Retina and Vitreous, Aravind Eye Hospital; Department of Proteomics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Prithviraj Udaya
- Department of Retina and Vitreous, Aravind Eye Hospital; Department of Proteomics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Jayapal J Maheshwari
- Department of Proteomics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Kim Ramasamy
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Naresh B Kannan
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | | |
Collapse
|
9
|
Das L, Shekhar S, Chandrani P, Varma AK. In silico structural analysis of secretory clusterin to assess pathogenicity of mutations identified in the evolutionarily conserved regions. J Biomol Struct Dyn 2023; 41:469-478. [PMID: 34821197 DOI: 10.1080/07391102.2021.2007791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Clusterin (CLU) is a secreted glycoprotein, heterodimeric in nature, and is expressed in a wide variety of tissues and body fluids such as serum and plasma. CLU has also been known to be a promising biomarker for cell death, malignancy, cancer progression, and resistance development. However, the lack of a CLU crystal structure obstructs understanding the possible role of reported mutations on the structure, and the subsequent effects on downstream signaling pathways and cancer progression. Considering the importance of crystal structure, a model structure of the pre-secretory isoform of CLU was built to predict the effect of mutations at the molecular level. Ab initio model was built using RaptorX, and loop refinement and energy minimization were carried out with ModLoop, ModRefiner, and GalaxyWeb servers. The cancer associated mutational spectra of CLU was retrieved from the cBioPortal server and 117 unique missense mutations were identified. Evolutionarily conserved regions and pathogenicity of mutations identified in CLU were analyzed using ConSurf and Rhapsody, respectively. Furthermore, sequence and structure-based mutational analysis were carried out with iSTABLE, DynaMut and PremPS servers. Molecular dynamics simulations were carried out with GROMACS for 50 ns to determine the stability of the wild type and mutant protein structures. A dynamically stable model structure of pre-secretory CLU (psCLU) which has high concurrence with the sequence based secondary structure predictions has been explored. Changes in the intra-atomic interactions and folding pattern between wild type and mutant structures were observed. To our conclusion, eleven mutations with the highest structural and functional significance have been predicted to have pathogenic and deleterious effects.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Lipi Das
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Shashank Shekhar
- Supercomputing Facility for Bioinformatics & Computational Biology, Indian Institute of Technology, New Delhi, India
| | - Pratik Chandrani
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India.,Medical Oncology Molecular Lab, Tata Memorial Hospital, Mumbai, India
| | - Ashok K Varma
- Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.,Homi Bhabha National Institute, Training School Complex, Mumbai, India
| |
Collapse
|
10
|
Endoplasmic Reticulum Stress-Regulated Chaperones as a Serum Biomarker Panel for Parkinson's Disease. Mol Neurobiol 2023; 60:1476-1485. [PMID: 36478320 PMCID: PMC9899193 DOI: 10.1007/s12035-022-03139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Examination of post-mortem brain tissues has previously revealed a strong association between Parkinson's disease (PD) pathophysiology and endoplasmic reticulum (ER) stress. Evidence in the literature regarding the circulation of ER stress-regulated factors released from neurons provides a rationale for investigating ER stress biomarkers in the blood to aid diagnosis of PD. The levels of ER stress-regulated proteins in serum collected from 29 PD patients and 24 non-PD controls were measured using enzyme-linked immunosorbent assays. A panel of four biomarkers, protein disulfide-isomerase A1, protein disulfide-isomerase A3, mesencephalic astrocyte-derived neurotrophic factor, and clusterin, together with age and gender had higher ability (area under the curve 0.64, sensitivity 66%, specificity 57%) and net benefit to discriminate PD patients from the non-PD group compared with other analyzed models. Addition of oligomeric and total α-synuclein to the model did not improve the diagnostic power of the biomarker panel. We provide evidence that ER stress-regulated proteins merit further investigation for their potential as diagnostic biomarkers of PD.
Collapse
|
11
|
Milinkeviciute G, Green KN. Clusterin/apolipoprotein J, its isoforms and Alzheimer's disease. Front Aging Neurosci 2023; 15:1167886. [PMID: 37122381 PMCID: PMC10133478 DOI: 10.3389/fnagi.2023.1167886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Affiliation(s)
- Giedre Milinkeviciute
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Giedre Milinkeviciute
| | - Kim N. Green
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
12
|
Timofeeva AV, Fedorov IS, Tarasova AM, Gorina KA, Suhova Y, Gusar VA, Ivanets TY. Role of clusterin in predicting development of early- and late-onset preeclampsia in the first trimester of pregnancy. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2022. [DOI: 10.24075/brsmu.2022.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Preeclampsia (PE) occurs in 2–8% of pregnancies. It is one of the leading causes of maternal and perinatal morbidity and mortality. Today, there are no tests adopted by the practitioners that enable accurate prediction of early (weeks 20 through 34) or late (after week 34) onset of PE when the pregnancy is in its 11th to 14th week. This study aimed to evaluate the feasibility of using secretory clusterin quantification to predict early or late PE during the first trimester of pregnancy. The choice of this protein is determined, on the one hand, by the specificity of its expression for cytotrophoblast, syncytiotrophoblast, and extracellular trophoblast cells, and, on the other hand, by the proven negative effect of clusterin on the invasive properties of trophoblastic cells and gestational transformations of uterine vessels, which play a key role in the pathogenesis of PE. The study included 40 pregnant women aged 27–40 years who underwent a comprehensive screening examination in the first trimester of pregnancy. Western blotting revealed a significant increase in the level of secretory clusterin (40 kDa) in the blood serum of pregnant women in the case of PE compared to physiological pregnancy: in early-onset PE, a twofold increase in the level of clusterin in the vesicular and extravesicular fractions of blood serum (p = 0.03 and p = 0.004, respectively), with late-onset PE — a threefold increase only in the extravesicular fraction of blood serum (p = 0.002). According to logistic regression models, the level of secretory clusterin in the extravesicular fraction of blood serum of pregnant women in the first trimester has prognostic significance in assessing the likelihood of developing early-onset PE (AUC = 0.97, Se = 1, Sp = 0.875, cutoff = 0.3877) and late-onset PE ( AUC = 1, Se = 1, Sp = 1, cutoff = 0.5).
Collapse
Affiliation(s)
- AV Timofeeva
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - IS Fedorov
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - AM Tarasova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - KA Gorina
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - YuV Suhova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - VA Gusar
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - TYu Ivanets
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| |
Collapse
|
13
|
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
|
14
|
The Influence of Clusterin Glycosylation Variability on Selected Pathophysiological Processes in the Human Body. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7657876. [PMID: 36071866 PMCID: PMC9441386 DOI: 10.1155/2022/7657876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022]
Abstract
The present review gathers together the most important information about variability in clusterin molecular structure, its profile, and the degree of glycosylation occurring in human tissues and body fluids in the context of the utility of these characteristics as potential diagnostic biomarkers of selected pathophysiological conditions. The carbohydrate part of clusterin plays a crucial role in many biological processes such as endocytosis and apoptosis. Many pathologies associated with neurodegeneration, carcinogenesis, metabolic diseases, and civilizational diseases (e.g., cardiovascular incidents and male infertility) have been described as causes of homeostasis disturbance, in which the glycan part of clusterin plays a very important role. The results of the discussed studies suggest that glycoproteomic analysis of clusterin may help differentiate the severity of hippocampal atrophy, detect the causes of infertility with an immune background, and monitor the development of cancer. Understanding the mechanism of clusterin (CLU) action and its binding epitopes may enable to indicate new therapeutic goals. The carbohydrate part of clusterin is considered necessary to maintain its proper molecular conformation, structural stability, and proper systemic and/or local biological activity. Taking into account the wide spectrum of CLU action and its participation in many processes in the human body, further studies on clusterin glycosylation variability are needed to better understand the molecular mechanisms of many pathophysiological conditions. They can also provide the opportunity to find new biomarkers and enrich the panel of diagnostic parameters for diseases that still pose a challenge for modern medicine.
Collapse
|
15
|
Wilson MR, Satapathy S, Jeong S, Fini ME. Clusterin, other extracellular chaperones, and eye disease. Prog Retin Eye Res 2022; 89:101032. [PMID: 34896599 PMCID: PMC9184305 DOI: 10.1016/j.preteyeres.2021.101032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
Proteostasis refers to all the processes that maintain the correct expression level, location, folding and turnover of proteins, essential to organismal survival. Both inside cells and in body fluids, molecular chaperones play key roles in maintaining proteostasis. In this article, we focus on clusterin, the first-recognized extracellular mammalian chaperone, and its role in diseases of the eye. Clusterin binds to and inhibits the aggregation of proteins that are misfolded due to mutations or stresses, clears these aggregating proteins from extracellular spaces, and facilitates their degradation. Clusterin exhibits three main homeostatic activities: proteostasis, cytoprotection, and anti-inflammation. The so-called "protein misfolding diseases" are caused by aggregation of misfolded proteins that accumulate pathologically as deposits in tissues; we discuss several such diseases that occur in the eye. Clusterin is typically found in these deposits, which is interpreted to mean that its capacity as a molecular chaperone to maintain proteostasis is overwhelmed in the disease state. Nevertheless, the role of clusterin in diseases involving such deposits needs to be better defined before therapeutic approaches can be entertained. A more straightforward case can be made for therapeutic use of clusterin based on its proteostatic role as a proteinase inhibitor, as well as its cytoprotective and anti-inflammatory properties. It is likely that clusterin works together in this way with other extracellular chaperones to protect the eye from disease, and we discuss several examples. We end this article by predicting future steps that may lead to development of clusterin as a biological drug.
Collapse
Affiliation(s)
- Mark R Wilson
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Sandeep Satapathy
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Shinwu Jeong
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1333 San Pablo Street., Los Angeles, CA, 90033, USA.
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine; Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences, Tufts University, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
16
|
Kremlitzka M, Colineau L, Nowacka AA, Mohlin FC, Wozniak K, Blom AM, King BC. Alternative translation and retrotranslocation of cytosolic C3 that detects cytoinvasive bacteria. Cell Mol Life Sci 2022; 79:291. [PMID: 35546365 PMCID: PMC9095555 DOI: 10.1007/s00018-022-04308-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/29/2022] [Accepted: 04/12/2022] [Indexed: 01/05/2023]
Abstract
Complement C3 was originally regarded as a serum effector protein, although recent data has emerged suggesting that intracellular C3 can also regulate basic cellular processes. Despite the growing interest in intracellular C3 functions, the mechanism behind its generation has not been demonstrated. In this study we show that C3 can be expressed from an alternative translational start site, resulting in C3 lacking the signal peptide, which is therefore translated in the cytosol. In contrast to the secreted form, alternatively translated cytosolic C3 is not glycosylated, is present mainly in a reduced state, and is turned over by the ubiquitin–proteasome system. C3 can also be retrotranslocated from the endoplasmic reticulum into the cytosol, structurally resembling secreted C3. Finally, we demonstrate that intracellular cytosolic C3 can opsonize invasive Staphylococcus aureus within epithelial cell, slowing vacuolar escape as well as impacting bacterial survival on subsequent exposure to phagocytes. Our work therefore reveals the existence and origin of intracellular, cytosolic C3, and demonstrates functions for cytosolic C3 in intracellular detection of cytoinvasive pathogens.
Collapse
Affiliation(s)
- Mariann Kremlitzka
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Lucie Colineau
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Alicja A Nowacka
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Frida C Mohlin
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Katarzyna Wozniak
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden.
| | - Ben C King
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
17
|
Yuste-Checa P, Bracher A, Hartl FU. The chaperone Clusterin in neurodegeneration-friend or foe? Bioessays 2022; 44:e2100287. [PMID: 35521968 DOI: 10.1002/bies.202100287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022]
Abstract
Fibrillar protein aggregates are the pathological hallmark of a group of age-dependent neurodegenerative conditions, including Alzheimer's and Parkinson's disease. Aggregates of the microtubule-associated protein Tau are observed in Alzheimer's disease and primary tauopathies. Tau pathology propagates from cell to cell in a prion-like process that is likely subject to modulation by extracellular chaperones such as Clusterin. We recently reported that Clusterin delayed Tau fibril formation but enhanced the activity of Tau oligomers to seed aggregation of endogenous Tau in a cellular model. In contrast, Clusterin inhibited the propagation of α-Synuclein aggregates associated with Parkinson's disease. These findings raise the possibility of a mechanistic link between Clusterin upregulation observed in Alzheimer's disease and the progression of Tau pathology. Here we review the diverse functions of Clusterin in the pathogenesis of neurodegenerative diseases, focusing on evidence that Clusterin may act either as a suppressor or enhancer of pathology.
Collapse
Affiliation(s)
- Patricia Yuste-Checa
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
18
|
Mesgarzadeh JS, Buxbaum JN, Wiseman RL. Stress-responsive regulation of extracellular proteostasis. J Cell Biol 2022; 221:213026. [PMID: 35191945 PMCID: PMC8868021 DOI: 10.1083/jcb.202112104] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/18/2022] Open
Abstract
Genetic, environmental, and aging-related insults can promote the misfolding and subsequent aggregation of secreted proteins implicated in the pathogenesis of numerous diseases. This has led to considerable interest in understanding the molecular mechanisms responsible for regulating proteostasis in extracellular environments such as the blood and cerebrospinal fluid (CSF). Extracellular proteostasis is largely dictated by biological pathways comprising chaperones, folding enzymes, and degradation factors localized to the ER and extracellular space. These pathways limit the accumulation of nonnative, potentially aggregation-prone proteins in extracellular environments. Many reviews discuss the molecular mechanisms by which these pathways impact the conformational integrity of the secreted proteome. Here, we instead focus on describing the stress-responsive mechanisms responsible for adapting ER and extracellular proteostasis pathways to protect the secreted proteome from pathologic insults that challenge these environments. Further, we highlight new strategies to identify stress-responsive pathways involved in regulating extracellular proteostasis and describe the pathologic and therapeutic implications for these pathways in human disease.
Collapse
Affiliation(s)
| | - Joel N Buxbaum
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | - R Luke Wiseman
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| |
Collapse
|
19
|
Kalvaityte U, Matta C, Bernotiene E, Pushparaj PN, Kiapour AM, Mobasheri A. Exploring the translational potential of clusterin as a biomarker of early osteoarthritis. J Orthop Translat 2022; 32:77-84. [PMID: 34976733 PMCID: PMC8671091 DOI: 10.1016/j.jot.2021.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Clusterin (CLU; also known as apolipoprotein J) is an ATP-independent holdase chaperone that prevents proteotoxicity as a consequence of protein aggregation. It is a ∼60 kDa disulfide-linked heterodimeric protein involved in the clearance of cellular debris and the regulation of apoptosis. CLU has been proposed to protect cells from cytolysis by complement components and has been implicated in Alzheimer's disease due to its ability to bind amyloid-β peptides and prevent aggregate formation in the brain. Recent studies suggest that CLU performs moonlighting functions. CLU exists in two major forms: an intracellular form and a secreted extracellular form. The intracellular form of CLU may suppress stress-induced apoptosis by forming complexes with misfolded proteins and facilitates their degradation. The secreted form of CLU functions as an extracellular chaperone that prevents protein aggregation. METHODS In this review, we discuss the published literature on the biology of CLU in cartilage, chondrocytes, and other synovial joint tissues. We also review clinical studies that have examined the potential for using this protein as a biomarker in synovial and systemic fluids of patients with rheumatoid arthritis (RA) or osteoarthritis (OA). RESULTS Since CLU functions as an extracellular chaperone, we propose that it may be involved in cytoprotective functions in osteoarticular tissues. The secreted form of CLU can be measured in synovial and systemic fluids and may have translational potential as a biomarker of early repair responses in OA. CONCLUSION There is significant potential for investigating synovial and systemic CLU as biomarkers of OA. Future translational and clinical orthopaedic studies should carefully consider the diverse roles of this protein and its involvement in other comorbidities. Therefore, future biomarker studies should not correlate circulating CLU levels exclusively to the process of OA pathogenesis and progression. Special attention should be paid to CLU levels in synovial fluid. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE There is significant potential for investigating synovial and systemic CLU as a predictive biomarker of osteoarthritis (OA) progression and response to novel treatments and interventions. Given that CLU plays diverse roles in other comorbidities such as rheumatoid arthritis (RA) and obesity, future translational and clinical orthopaedic biomarker studies should not directly correlate circulating CLU levels to the process of OA pathogenesis and progression. However, special attention should be paid to CLU levels in synovial fluid. The cytoprotective properties of CLU may support the implementation of regenerative strategies and new approaches for developing targeted therapeutics for OA.
Collapse
Key Words
- ACL, anterior cruciate ligament
- ACR, American College of Rheumatology
- ApoJ, apolipoprotein J
- Apoptosis
- CLU, clusterin
- CMC-I, carpometacarpal joint
- COMP, cartilage oligomeric matrix protein
- Clusterin (CLU)
- ECM, extracellular matrix
- ELISA, enzyme-linked immunosorbent assay
- ESCEO, The European Society for Clinical and Economic Aspects of Osteoporosis: Osteoarthritis and Musculoskeletal Diseases
- Inflammation
- OA, osteoarthritis
- OARSI, Osteoarthritis Research Society International
- Osteoarthritis (OA)
- PsA, psoriatic arthritis
- RA, rheumatoid arthritis
- Rheumatoid arthritis (RA)
- SF, synovial fluid
- TNF-α, tumor necrosis factor-α
- Translational biomarker
- hsCRP, high sensitivity C-reactive protein
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
- sCLU, secreted clusterin
Collapse
Affiliation(s)
- Ursule Kalvaityte
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, H, 4032, Hungary
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ata M. Kiapour
- Department of Orthopedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 021115, USA
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT, 08406, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI, 90014, Oulu, Finland
- Department of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508, GA, Utrecht, the Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
20
|
Wei B, Zhu Y, Yang P, Han Y, Wang S, Wang X, Xia S, Song X, Zhang Z, Wang S, Rondard P, Pin JP, Jiang X, Liu J. GABA B1e promotes the malignancy of human cancer cells by targeting the tyrosine phosphatase PTPN12. iScience 2021; 24:103311. [PMID: 34778730 PMCID: PMC8577127 DOI: 10.1016/j.isci.2021.103311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/24/2021] [Accepted: 10/14/2021] [Indexed: 01/30/2023] Open
Abstract
Neurotransmitter receptors are involved in cancer progression. Among them, the heterodimeric GABAB receptor, activated by the main inhibitory neurotransmitter GABA, is composed of the transmembrane GABAB1 and GABAB2 subunits. The oncogenic role of the isoform GABAB1e (GB1e) containing only the extracellular domain of GABAB1 remains unclear. We revealed that GB1e is largely expressed in human breast cancer (BrCa) cell lines as well as in BrCa tissues where it is upregulated. Moreover, GB1e promoted the malignancy of BrCa cells both in vitro and in vivo. We propose that GB1e favors EGFR signaling by interacting with PTPN12 to disrupt the interaction between EGFR and PTPN12, and phosphorylation of Y230 and Y404 on GB1e is required in this process. Our data highlight that the GABBR1 gene through the expression of the GB1e isoform might play an important oncogenic role in BrCa and that GB1e is of interest for the treatment of some cancers. GABAB1e promotes the malignancy of breast cancer cells both in vitro and in vivo Specific phosphorylation of GABAB1e is critical for its association with PTPN12 GABAB1e disrupts EGFR interacting with PTPN12 and induces EGFR-PI3K/Akt signaling
Collapse
Affiliation(s)
- Bo Wei
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Yini Zhu
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Peng Yang
- Department of Breast & Endocrine Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China
| | - Yong Han
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China
| | - Suyun Wang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Xiaomei Wang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Shuai Xia
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Xiaoguang Song
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Zhongling Zhang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Sheng Wang
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Xinnong Jiang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
- Corresponding author
| | - Jianfeng Liu
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
- Corresponding author
| |
Collapse
|
21
|
Sun HY, Chen TY, Tan YC, Wang CH, Young KC. Sterol O-acyltransferase 2 chaperoned by apolipoprotein J facilitates hepatic lipid accumulation following viral and nutrient stresses. Commun Biol 2021; 4:564. [PMID: 33980978 PMCID: PMC8115332 DOI: 10.1038/s42003-021-02093-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 04/06/2021] [Indexed: 11/08/2022] Open
Abstract
The risks of non-alcoholic fatty liver disease (NAFLD) include obese and non-obese stresses such as chronic hepatitis C virus (HCV) infection, but the regulatory determinants remain obscure. Apolipoprotein J (ApoJ) served as an ER-Golgi contact-site chaperone near lipid droplet (LD), facilitating HCV virion production. We hypothesized an interplay between hepatic ApoJ, cholesterol esterification and lipid deposit in response to NAFLD inducers. Exposures of HCV or free-fatty acids exhibited excess LDs along with increased ApoJ expression, whereas ApoJ silencing alleviated hepatic lipid accumulation. Both stresses could concomitantly disperse Golgi, induce closer ApoJ and sterol O-acyltransferase 2 (SOAT2) contacts via the N-terminal intrinsically disordered regions, and increase cholesteryl-ester. Furthermore, serum ApoJ correlated positively with cholesterol and low-density lipoprotein levels in normal glycaemic HCV patients, NAFLD patients and in mice with steatosis. Taken together, hepatic ApoJ might activate SOAT2 to supply cholesteryl-ester for lipid loads, thus providing a therapeutic target of stress-induced steatosis.
Collapse
Affiliation(s)
- Hung-Yu Sun
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, China
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Ying Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ching Tan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Hsiang Wang
- Division of Gastroenterology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Kung-Chia Young
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Rodríguez-Rivera C, Pérez-Carrión MD, Olavarría LC, Alguacil LF, Mora MJP, González-Martín C. Clusterin levels in undernourished SH-SY5Y cells. Food Nutr Res 2021; 65:5709. [PMID: 33994910 PMCID: PMC8098648 DOI: 10.29219/fnr.v65.5709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 11/20/2022] Open
Abstract
Food-related disorders are increasingly common in developed societies, and the psychological component of these disorders has been gaining increasing attention. Both overnourishment with high-fat diets and perinatal undernourishment in mice have been linked to a higher motivation toward food, resulting in an alteration in food intake. Clusterin (CLU), a multifaced protein, is overexpressed in the nucleus accumbens (NAc) of over-fed rats, as well as in those that suffered chronic undernutrition. Moreover, an increase of this protein was observed in the plasma of obese patients with food addiction, suggesting the implication of CLU in this eating disorder. To characterize CLU’s cellular mechanisms, in vitro experiments of undernutrition were performed using dopaminergic SH-SY5Y cells. To mimic in vivo dietary conditions, cells were treated with different fetal bovine serum (FBS) concentrations, resulting in control (C group) diet (10% FBS), undernourishment (U group) diet (0.5% FBS), and undernourishment diet followed by restoration of control diet (UC group) (0.5 + 10% FBS). Undernourishment compromised cell viability and proliferation, and concomitantly increased CLU secretion as well as the cytosolic pool of the protein, while decreasing the mitochondrial level. The restoration of normal conditions tended to recover cell physiology, and the normal levels and distribution of CLU. This research study is a step forward toward the characterization of clusterin as a potential marker for food addiction and nutritional status.
Collapse
Affiliation(s)
| | - María Dolores Pérez-Carrión
- Facultad de Farmacia, Universidad CEU San Pablo, Alcorcón, Madrid, Spain.,Facultad de Medicina, Universidad de Castilla-la Mancha, Albacete, Spain
| | | | - Luis F Alguacil
- Facultad de Farmacia, Universidad CEU San Pablo, Alcorcón, Madrid, Spain.,Facultad de Farmacia, Instituto de Estudio de las Adicciones, Universidad CEU San Pablo, Alcorcón, Madrid, Spain
| | - María José Polanco Mora
- Facultad de Farmacia, Universidad CEU San Pablo, Alcorcón, Madrid, Spain.,Facultad de Farmacia, Instituto de Estudio de las Adicciones, Universidad CEU San Pablo, Alcorcón, Madrid, Spain
| | - Carmen González-Martín
- Facultad de Farmacia, Universidad CEU San Pablo, Alcorcón, Madrid, Spain.,Facultad de Farmacia, Instituto de Estudio de las Adicciones, Universidad CEU San Pablo, Alcorcón, Madrid, Spain
| |
Collapse
|
23
|
Satapathy S, Wilson MR. The Dual Roles of Clusterin in Extracellular and Intracellular Proteostasis. Trends Biochem Sci 2021; 46:652-660. [PMID: 33573881 DOI: 10.1016/j.tibs.2021.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Clusterin (CLU) was the first reported secreted mammalian chaperone and impacts on serious diseases associated with inappropriate extracellular protein aggregation. Many studies have described intracellular CLU in locations outside the secretory system and recent work has shown that CLU can be released into the cytosol during cell stress. In this article, we critically evaluate evidence relevant to the proposed origins of cellular CLU found outside the secretory system, and advance the hypothesis that the cytosolic release of CLU induced by stress serves to facilitate the trafficking of misfolded proteins to the proteasome and autophagy for degradation. We also propose future research directions that could help establish CLU as a unique chaperone performing critical and synergic roles in both intracellular and extracellular proteostasis.
Collapse
Affiliation(s)
- Sandeep Satapathy
- School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Mark R Wilson
- School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| |
Collapse
|
24
|
Rodríguez-Rivera C, Garcia MM, Molina-Álvarez M, González-Martín C, Goicoechea C. Clusterin: Always protecting. Synthesis, function and potential issues. Biomed Pharmacother 2021; 134:111174. [DOI: 10.1016/j.biopha.2020.111174] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
|
25
|
Layalle S, They L, Ourghani S, Raoul C, Soustelle L. Amyotrophic Lateral Sclerosis Genes in Drosophila melanogaster. Int J Mol Sci 2021; 22:ijms22020904. [PMID: 33477509 PMCID: PMC7831090 DOI: 10.3390/ijms22020904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating adult-onset neurodegenerative disease characterized by the progressive degeneration of upper and lower motoneurons. Most ALS cases are sporadic but approximately 10% of ALS cases are due to inherited mutations in identified genes. ALS-causing mutations were identified in over 30 genes with superoxide dismutase-1 (SOD1), chromosome 9 open reading frame 72 (C9orf72), fused in sarcoma (FUS), and TAR DNA-binding protein (TARDBP, encoding TDP-43) being the most frequent. In the last few decades, Drosophila melanogaster emerged as a versatile model for studying neurodegenerative diseases, including ALS. In this review, we describe the different Drosophila ALS models that have been successfully used to decipher the cellular and molecular pathways associated with SOD1, C9orf72, FUS, and TDP-43. The study of the known fruit fly orthologs of these ALS-related genes yielded significant insights into cellular mechanisms and physiological functions. Moreover, genetic screening in tissue-specific gain-of-function mutants that mimic ALS-associated phenotypes identified disease-modifying genes. Here, we propose a comprehensive review on the Drosophila research focused on four ALS-linked genes that has revealed novel pathogenic mechanisms and identified potential therapeutic targets for future therapy.
Collapse
Affiliation(s)
- Sophie Layalle
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Laetitia They
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Sarah Ourghani
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence: (C.R.); (L.S.)
| | - Laurent Soustelle
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
- Correspondence: (C.R.); (L.S.)
| |
Collapse
|
26
|
Identification of novel substrates of a disintegrin and metalloprotease 17 by specific labeling of surface proteins. Heliyon 2021; 6:e05804. [PMID: 33385093 PMCID: PMC7770542 DOI: 10.1016/j.heliyon.2020.e05804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/05/2020] [Accepted: 12/18/2020] [Indexed: 11/22/2022] Open
Abstract
A disintegrin and metalloprotease 17 (ADAM17) catalyzes the cleavage and release of the ectodomains of its substrates at the cell surface in a process termed ectodomain shedding. However, not all ADAM17 substrates have been identified. Here, we used cell surface protein-specific labeling and proteomic approaches to detect and identify ADAM17 substrates. HeLa cell surface proteins were labeled with a fluorescent dye and cultured with or without TAPI-2, an ADAM17 inhibitor. Labeled proteins released into the culture medium were detected by 2-dimensional gel electrophoresis (2DE). Protein spots showing decreased intensity in response to TAPI-2 were selected as substrates of ADAM17 or their binding proteins, and identified by mass spectrometry. ADAM17 knockdown was preformed to examine the behavior of identified proteins. Of 347 proteins detected by 2DE, 49 showed lower intensity in TAPI-2 (+) than in TAPI-2 (-) samples (p < 0.05), and were considered as candidate substrates of ADAM17. Mass spectrometric analysis of 14 protein spots showing >50% decreased intensity identified clusterin as a novel ADAM17 substrate, in addition to known substrates such as desmoglein-2. Western blot analysis showed that ADAM17 knockdown decreased the levels of clusterin fragments cleaved and released from the cell surface. The results identified clusterin as a novel ADAM17 substrate. The method used to identify clusterin could be used to identify the substrates of other sheddases involved in ectodomain shedding.
Collapse
|
27
|
Intracellular leucine-rich alpha-2-glycoprotein-1 competes with Apaf-1 for binding cytochrome c in protecting MCF-7 breast cancer cells from apoptosis. Apoptosis 2021; 26:71-82. [PMID: 33386492 DOI: 10.1007/s10495-020-01647-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
Leucine-rich alpha-2-glycoprotein-1 (LRG1) has been shown to compete with apoptosis activating factor-1 (Apaf-1) for binding cytochrome c (Cyt c) and could play a role in inhibition of apoptosis. Employing MCF-7 breast cancer cells, we report that intracellular LRG1 does protect against apoptosis. Thus, cells transfected with the lrg1 gene and expressing higher levels of LRG1 were more resistant to hydrogen peroxide-induced apoptosis than parental cells, while cells in which LRG mRNA was knocked down by short hairpin (sh) RNA-induced degradation were more sensitive. The amount of Cyt c co-immunoprecipitated with Apaf-1 from the cytosol of apoptotic cells was inversely related to the level of LRG1 expression. In lrg1-transfected cells partially-glycosylated LRG1 was found in the cytosol and there was an increase in cytosolic Cyt c in live lrg1-transfected cells relative to parental cells. However, apoptosis was not spontaneously induced because Cyt c was bound to LRG1 and not to Apaf-1. Cyt c was the only detectable protein co-immunoprecipitated with LRG1. Following hydrogen peroxide treatment degradation of LRG1 allowed for induction of apoptosis. We propose that intracellular LRG1 raises the threshold of cytoplasmic Cyt c required to induce apoptosis and, thus, prevents onset of the intrinsic pathway in cells where Cyt c release from mitochondria does not result from committed apoptotic signaling. This mechanism of survival afforded by LRG1 is likely to be distinct from its extracellular survival function that has been reported by several research groups.
Collapse
|
28
|
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
|
29
|
Shakya M, Yildirim T, Lindberg I. Increased expression and retention of the secretory chaperone proSAAS following cell stress. Cell Stress Chaperones 2020; 25:929-941. [PMID: 32607937 PMCID: PMC7591655 DOI: 10.1007/s12192-020-01128-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 11/25/2022] Open
Abstract
The secretory pathway of neurons and endocrine cells contains a variety of mechanisms designed to combat cellular stress. These include not only the unfolded protein response pathways but also diverse chaperone proteins that collectively work to ensure proteostatic control of secreted and membrane-bound molecules. One of the least studied of these chaperones is the neural- and endocrine-specific molecule known as proSAAS. This small chaperone protein acts as a potent anti-aggregant both in vitro and in cellulo and also represents a cerebrospinal fluid biomarker in Alzheimer's disease. In the present study, we have examined the idea that proSAAS, like other secretory chaperones, might represent a stress-responsive protein. We find that exposure of neural and endocrine cells to the cell stressors tunicamycin and thapsigargin increases cellular proSAAS mRNA and protein in Neuro2A cells. Paradoxically, proSAAS secretion is inhibited by these same drugs. Exposure of Neuro2A cells to low concentrations of the hypoxic stress inducer cobalt chloride, or to sodium arsenite, an oxidative stressor, also increases cellular proSAAS content and reduces its secretion. We conclude that the cellular levels of the small secretory chaperone proSAAS are positively modulated by cell stress.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA
| | - Taha Yildirim
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA.
| |
Collapse
|
30
|
Yammine L, Zablocki A, Baron W, Terzi F, Gallazzini M. Lipocalin-2 Regulates Epidermal Growth Factor Receptor Intracellular Trafficking. Cell Rep 2020; 29:2067-2077.e6. [PMID: 31722218 DOI: 10.1016/j.celrep.2019.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 08/02/2019] [Accepted: 10/03/2019] [Indexed: 11/27/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) activation and lipocalin-2 (Lcn2) expression are frequently observed in the same pathological contexts, such as cancers or chronic kidney disease (CKD). However, the significance of this association is unknown. Here, we describe the role of Lcn2 in regulating EGFR trafficking. We show that Lcn2 increases EGFR cell surface abundance and is required for transforming growth factor α (TGF-α)-induced EGFR recycling to the plasma membrane and sustained activation. Lcn2 binds to the intracellular domain of EGFR in late endosomal compartments and inhibits its lysosomal degradation. Consistently, Lcn2 enhances EGFR-induced cell migration after TGF-α stimulation. In vivo, Lcn2 gene inactivation prevents EGFR recycling to the plasma membrane in an experimental model of CKD. Remarkably, this is associated with a dramatic decrease of renal lesions. Together, our data identify Lcn2 as a key mediator of EGFR trafficking processes. Hence, therapeutic inhibition of Lcn2 may counteract the deleterious effect of EGFR activation.
Collapse
Affiliation(s)
- Lucie Yammine
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Aniela Zablocki
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - William Baron
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Fabiola Terzi
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Morgan Gallazzini
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France.
| |
Collapse
|
31
|
ER Stress-Induced Secretion of Proteins and Their Extracellular Functions in the Heart. Cells 2020; 9:cells9092066. [PMID: 32927693 PMCID: PMC7563782 DOI: 10.3390/cells9092066] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a result of conditions that imbalance protein homeostasis or proteostasis at the ER, for example ischemia, and is a common event in various human pathologies, including the diseased heart. Cardiac integrity and function depend on the active secretion of mature proteins from a variety of cell types in the heart, a process that requires an intact ER environment for efficient protein folding and trafficking to the secretory pathway. As a consequence of ER stress, most protein secretion by the ER secretory pathway is decreased. Strikingly, there is a select group of proteins that are secreted in greater quantities during ER stress. ER stress resulting from the dysregulation of ER Ca2+ levels, for instance, stimulates the secretion of Ca2+-binding ER chaperones, especially GRP78, GRP94, calreticulin, and mesencephalic astrocyte-derived neurotrophic factor (MANF), which play a multitude of roles outside the cell, strongly depending on the cell type and tissue. Here we review current insights in ER stress-induced secretion of proteins, particularly from the heart, and highlight the extracellular functions of these proteins, ranging from the augmentation of cardiac cell viability to the modulation of pro- and anti-apoptotic, oncogenic, and immune-stimulatory cell signaling, cell invasion, extracellular proteostasis, and more. Many of the roles of ER stress-induced protein secretion remain to be explored in the heart. This article is part of a special issue entitled “The Role of Proteostasis Derailment in Cardiac Diseases.”
Collapse
|
32
|
Satapathy S, Dabbs RA, Wilson MR. Rapid high-yield expression and purification of fully post-translationally modified recombinant clusterin and mutants. Sci Rep 2020; 10:14243. [PMID: 32859921 PMCID: PMC7455699 DOI: 10.1038/s41598-020-70990-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/30/2020] [Indexed: 11/17/2022] Open
Abstract
The first described and best known mammalian secreted chaperone, abundant in human blood, is clusterin. Recent independent studies are now exploring the potential use of clusterin as a therapeutic in a variety of disease contexts. In the past, the extensive post-translational processing of clusterin, coupled with its potent binding to essentially any misfolded protein, have meant that its expression as a fully functional recombinant protein has been very difficult. We report here the first rapid and high-yield system for the expression and purification of fully post-translationally modified and chaperone-active clusterin. Only 5–6 days is required from initial transfection to harvest of the protein-free culture medium containing the recombinant product. Purification to near-homogeneity can then be accomplished in a single affinity purification step and the yield for wild type human clusterin is of the order of 30–40 mg per litre of culture. We have also shown that this system can be used to quickly express and purify custom-designed clusterin mutants. These advances dramatically increase the feasibility of detailed structure–function analysis of the clusterin molecule and will facilitate identification of those specific regions responsible for the interactions of clusterin with receptors and other molecules.
Collapse
Affiliation(s)
- Sandeep Satapathy
- Illawarra Health and Medical Research Institute, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.,Molecular Horizons Research Institute, University of Wollongong, Rm 313, Building 42 (Molecular Horizons), Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Rebecca A Dabbs
- Burnett Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia. .,Molecular Horizons Research Institute, University of Wollongong, Rm 313, Building 42 (Molecular Horizons), Northfields Avenue, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
33
|
Chaplot K, Jarvela TS, Lindberg I. Secreted Chaperones in Neurodegeneration. Front Aging Neurosci 2020; 12:268. [PMID: 33192447 PMCID: PMC7481362 DOI: 10.3389/fnagi.2020.00268] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis, or proteostasis, is a combination of cellular processes that govern protein quality control, namely, protein translation, folding, processing, and degradation. Disruptions in these processes can lead to protein misfolding and aggregation. Proteostatic disruption can lead to cellular changes such as endoplasmic reticulum or oxidative stress; organelle dysfunction; and, if continued, to cell death. A majority of neurodegenerative diseases involve the pathologic aggregation of proteins that subverts normal neuronal function. While prior reviews of neuronal proteostasis in neurodegenerative processes have focused on cytoplasmic chaperones, there is increasing evidence that chaperones secreted both by neurons and other brain cells in the extracellular - including transsynaptic - space play important roles in neuronal proteostasis. In this review, we will introduce various secreted chaperones involved in neurodegeneration. We begin with clusterin and discuss its identification in various protein aggregates, and the use of increased cerebrospinal fluid (CSF) clusterin as a potential biomarker and as a potential therapeutic. Our next secreted chaperone is progranulin; polymorphisms in this gene represent a known genetic risk factor for frontotemporal lobar degeneration, and progranulin overexpression has been found to be effective in reducing Alzheimer's- and Parkinson's-like neurodegenerative phenotypes in mouse models. We move on to BRICHOS domain-containing proteins, a family of proteins containing highly potent anti-amyloidogenic activity; we summarize studies describing the biochemical mechanisms by which recombinant BRICHOS protein might serve as a therapeutic agent. The next section of the review is devoted to the secreted chaperones 7B2 and proSAAS, small neuronal proteins which are packaged together with neuropeptides and released during synaptic activity. Since proteins can be secreted by both classical secretory and non-classical mechanisms, we also review the small heat shock proteins (sHsps) that can be secreted from the cytoplasm to the extracellular environment and provide evidence for their involvement in extracellular proteostasis and neuroprotection. Our goal in this review focusing on extracellular chaperones in neurodegenerative disease is to summarize the most recent literature relating to neurodegeneration for each secreted chaperone; to identify any common mechanisms; and to point out areas of similarity as well as differences between the secreted chaperones identified to date.
Collapse
Affiliation(s)
| | | | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
34
|
Holmannova D, Borsky P, Borska L, Andrys C, Hamakova K, Rehacek V, Svadlakova T, Malkova A, Beranek M, Palicka V, Krejsek J, Fiala Z. Metabolic Syndrome, Clusterin and Elafin in Patients with Psoriasis Vulgaris. Int J Mol Sci 2020; 21:ijms21165617. [PMID: 32764517 PMCID: PMC7460615 DOI: 10.3390/ijms21165617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Psoriasis is a pathological condition characterized by immune system dysfunction and inflammation. Patients with psoriasis are more likely to develop a wide range of disorders associated with inflammation. Serum levels of various substances and their combinations have been associated with the presence of the disease (psoriasis) and have shown the potential to reflect its activity. The aim of the present study is to contribute to the elucidation of pathophysiological links between psoriasis, its pro-inflammatory comorbidity metabolic syndrome (MetS), and the expression of clusterin and elafin, which are reflected in the pathophysiological “portfolio” of both diseases. Material and methods: Clinical examinations (PASI score), ELISA (clusterin, elafin), and biochemical analyses (parameters of MetS) were performed. Results: We found that patients with psoriasis were more often afflicted by MetS, compared to the healthy controls. Clusterin and elafin levels were higher in the patients than in the controls but did not correlate to the severity of psoriasis. Conclusion: Our data suggest that patients with psoriasis are more susceptible to developing other systemic inflammatory diseases, such as MetS. The levels of clusterin and elafin, which are tightly linked to inflammation, were significantly increased in the patients, compared to the controls, but the presence of MetS in patients did not further increase these levels.
Collapse
Affiliation(s)
- Drahomira Holmannova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| | - Pavel Borsky
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Pathological Physiology, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic;
- Correspondence:
| | - Lenka Borska
- Institute of Pathological Physiology, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic;
| | - Ctirad Andrys
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Kvetoslava Hamakova
- Clinic of Dermatology and Venereology, University Hospital Hradec Kralove, 50005 Hradec Králové, Czech Republic;
| | - Vit Rehacek
- Transfusion Center, University Hospital, 50005 Hradec Kralove, Czech Republic;
| | - Tereza Svadlakova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Andrea Malkova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| | - Martin Beranek
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic;
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic;
| | - Jan Krejsek
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Zdenek Fiala
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| |
Collapse
|
35
|
Early Elevation of Systemic Plasma Clusterin after Reperfused Acute Myocardial Infarction in a Preclinical Porcine Model of Ischemic Heart Disease. Int J Mol Sci 2020; 21:ijms21134591. [PMID: 32605184 PMCID: PMC7369988 DOI: 10.3390/ijms21134591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/14/2020] [Accepted: 06/23/2020] [Indexed: 01/08/2023] Open
Abstract
Clusterin exerts anti-inflammatory, cytoprotective and anti-apoptotic effects. Both an increase and decrease of clusterin in acute myocardial infarction (AMI) has been reported. We aimed to clarify the role of clusterin as a systemic biomarker in AMI. AMI was induced by percutaneous left anterior artery (LAD) occlusion for 90 min followed by reperfusion in 24 pigs. Contrast ventriculography was performed after reperfusion to assess left ventricular ejection fraction (LVEF), left ventricular end diastolic volume (LVEDV) and left ventricular end systolic volume (LVESV) and additional cMRI + late enhancement to measure infarct size and LV functions at day 3 and week 6 post-MI. Blood samples were collected at prespecified timepoints. Plasma clusterin and other biomarkers (cTnT, NT-proBNP, neprilysin, NGAL, ET-1, osteopontin, miR21, miR29) were measured by ELISA and qPCR. Gene expression profiles of infarcted and remote region 3 h (n = 5) and 3 days (n = 5) after AMI onset were analysed by RNA-sequencing. AMI led to an increase in LVEDV and LVESV during 6-week, with concomitant elevation of NT-proBNP 3-weeks after AMI. Plasma clusterin levels were increased immediately after AMI and returned to normal levels until 3-weeks. Plasma NGAL, ET-1 and miR29 was significantly elevated at 3 weeks follow-up, miR21 increased after reperfusion and at 3 weeks post-AMI, while circulating neprilysin levels did not change. Elevated plasma clusterin levels 120 min after AMI onset suggest that clusterin might be an additional early biomarker of myocardial ischemia.
Collapse
|
36
|
Whiten DR, Cox D, Horrocks MH, Taylor CG, De S, Flagmeier P, Tosatto L, Kumita JR, Ecroyd H, Dobson CM, Klenerman D, Wilson MR. Single-Molecule Characterization of the Interactions between Extracellular Chaperones and Toxic α-Synuclein Oligomers. Cell Rep 2019; 23:3492-3500. [PMID: 29924993 PMCID: PMC6024880 DOI: 10.1016/j.celrep.2018.05.074] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/26/2018] [Accepted: 05/22/2018] [Indexed: 12/28/2022] Open
Abstract
The aberrant aggregation of α-synuclein is associated with several human diseases, collectively termed the α-synucleinopathies, which includes Parkinson’s disease. The progression of these diseases is, in part, mediated by extracellular α-synuclein oligomers that may exert effects through several mechanisms, including prion-like transfer, direct cytotoxicity, and pro-inflammatory actions. In this study, we show that two abundant extracellular chaperones, clusterin and α2-macroglobulin, directly bind to exposed hydrophobic regions on the surface of α-synuclein oligomers. Using single-molecule fluorescence techniques, we found that clusterin, unlike α2-macroglobulin, exhibits differential binding to α-synuclein oligomers that may be related to structural differences between two previously described forms of αS oligomers. The binding of both chaperones reduces the ability of the oligomers to permeabilize lipid membranes and prevents an oligomer-induced increase in ROS production in cultured neuronal cells. Taken together, these data suggest a neuroprotective role for extracellular chaperones in suppressing the toxicity associated with α-synuclein oligomers. Two extracellular chaperones directly bind to α-synuclein oligomers The binding is mediated by hydrophobicity on the oligomer surface Bound chaperones significantly attenuate the toxicity of α-synuclein oligomers
Collapse
Affiliation(s)
- Daniel R Whiten
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong 2522, NSW, Australia
| | - Dezerae Cox
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong 2522, NSW, Australia
| | - Mathew H Horrocks
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong 2522, NSW, Australia
| | - Christopher G Taylor
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Suman De
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Patrick Flagmeier
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Laura Tosatto
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Janet R Kumita
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong 2522, NSW, Australia
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong 2522, NSW, Australia.
| |
Collapse
|
37
|
Clusterin Silencing in Prostate Cancer Induces Matrix Metalloproteinases by an NF- κB-Dependent Mechanism. JOURNAL OF ONCOLOGY 2019; 2019:4081624. [PMID: 31885575 PMCID: PMC6925831 DOI: 10.1155/2019/4081624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/31/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
Abstract
Clusterin (CLU) is a stress-activated glycoprotein, whose expression is altered both in inflammation and cancer. Previously, we showed that abrogation of CLU expression in cancer-prone mice (TRAMP) results in the enhancement of tumor spreading and homing, concomitant with an enhanced expression of NF-κB. In the present paper, we carried out an extensive experimental work by utilizing microarray gene expression data, as well as in vitro and in vivo models of prostate cancer (PCa). Our results demonstrated that (i) CLU expression is significantly downregulated in human PCa and inversely correlates with the expression of p65 in metastases; (ii) CLU overexpression in PCa cells reduces the Ser536 phosphorylation of p65, inhibits NF-κB nuclear translocation, and reduces the transcription of matrix metalloproteinase-9 and metalloproteinase-2 (MMP-9 and MMP-2). Conversely, CLU silencing promotes NF-κB activation and transcriptional upregulation of MMP-9; and (iii) expression and activity of MMP-2 and MMP-9 are increased in CLU−/− mice (CLUKO) and in TRAMP/CLUKO mice in comparison to their relative Clu+/+ littermates. Taken together, our data support the hypothesis that CLU downregulation, an early and relevant event in PCa onset, may inhibit NF-κB activation and limit the execution of a transcriptional program that favor the disease progression towards a metastatic stage.
Collapse
|
38
|
Gregory JM, Elliott E, McDade K, Bak T, Pal S, Chandran S, Abrahams S, Smith C. Neuronal clusterin expression is associated with cognitive protection in amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol 2019; 46:255-263. [PMID: 31386770 PMCID: PMC7318312 DOI: 10.1111/nan.12575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/12/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
Abstract
AIMS Clusterin is a topologically dynamic chaperone protein with the ability to participate in both intra- and extacellular proteostasis. Clusterin has been shown to be upregulated in the spinal cord of patients with amyotrophic lateral sclerosis (ALS) and has been shown to protect against TDP-43 protein misfolding in animal and cell models. Previous studies have demonstrated an association between the pathological burden of TDP-43 misfolding and cognitive deficits in ALS, demonstrating high specificity, but correspondingly low sensitivity owing to a subset of individuals with no evidence of cognitive deficits despite a high burden of TDP-43 pathology, called mismatch cases. METHODS Hypothesizing that differences in the ability to cope with protein misfolding in these cases may be due to differences in expression of protective mechanisms such as clusterin expression, we assessed the spatial expression of clusterin and another chaperone protein, HspB8, in post mortem brain tissue of mismatch cases. We employed a modified in situ hybridization technique called BaseScope, with single cell, single transcript resolution. RESULTS Mismatch cases demonstrated differential spatial expression of clusterin, with a predominantly neuronal pattern, compared to cases with cognitive manifestations of their TDP-43 pathology who demonstrated a predominantly glial distribution of expression. CONCLUSIONS Our data suggest that, in individuals with TDP-43 pathology, predominantly neuronal expression of clusterin in extra-motor brain regions may indicate a cell protective mechanism delaying clinical manifestations such as cognitive dysfunction.
Collapse
Affiliation(s)
- J M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - E Elliott
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - K McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - T Bak
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Human Cognitive Neuroscience, Psychology, University of Edinburgh, Edinburgh, UK
| | - S Pal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - S Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - S Abrahams
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Human Cognitive Neuroscience, Psychology, University of Edinburgh, Edinburgh, UK
| | - C Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
39
|
Iłżecka J, Iłżecki M, Grabarska A, Dave S, Feldo M, Zubilewicz T. Clusterin as a potential marker of brain ischemia-reperfusion injury in patients undergoing carotid endarterectomy. Ups J Med Sci 2019; 124:193-198. [PMID: 31460820 PMCID: PMC6758642 DOI: 10.1080/03009734.2019.1646359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Introduction: Carotid endarterectomy (CEA) is a surgical procedure used in the prevention of ischemic stroke. However, this procedure can cause complications of ischemia-reperfusion injury to the brain. Clusterin (CLU) is a cytoprotective chaperone protein that is released from neurons in response to various neurological injuries. The objective of the study was to report the changes in serum CLU concentrations of patients undergoing CEA. Materials and methods: The study involved 25 patients with severe internal carotid artery stenosis. Serum samples were taken from patients at three different times: within 24 hours preoperatively to CEA, 12 hours postoperatively, and 48 hours postoperatively. Serum CLU concentrations were measured using a commercially available enzyme-linked immunosorbent assay. Results: When compared to concentrations preoperatively, the serum CLU concentration initially decreased during the 12 hours following CEA. However, 48 hours following the procedure there was an increase in the CLU concentration. After statistical analysis, differences were detected in serum CLU concentration between all three recorded measurements (P < 0.05). Conclusion: Data from our study indicate that serum CLU concentrations are affected after CEA. We hypothesize that serum CLU concentrations may depend on brain ischemia-reperfusion injury following this surgical procedure.
Collapse
Affiliation(s)
- Joanna Iłżecka
- Independent Neurological Rehabilitation Unit, Medical University of Lublin, Lublin, Poland
- CONTACT Joanna Iłżecka, MD, PhD Independent Neurological Rehabilitation Unit, S. Staszica 4/6, 20-081 Lublin, Poland
| | - Marek Iłżecki
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Aneta Grabarska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Shawn Dave
- University of Oklahoma Health Sciences Center in Oklahoma City, Oklahoma, USA
| | - Marcin Feldo
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Tomasz Zubilewicz
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
40
|
Contu L, Carare RO, Hawkes CA. Knockout of apolipoprotein A-I decreases parenchymal and vascular β-amyloid pathology in the Tg2576 mouse model of Alzheimer's disease. Neuropathol Appl Neurobiol 2019; 45:698-714. [PMID: 31002190 DOI: 10.1111/nan.12556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 04/10/2019] [Indexed: 11/30/2022]
Abstract
AIMS Apolipoprotein A-I (apoA-I), the principal apolipoprotein associated with high-density lipoproteins in the periphery, is also found at high concentrations in the cerebrospinal fluid. Previous studies have reported either no impact or vascular-specific effects of apoA-I knockout (KO) on β-amyloid (Aβ) pathology. However, the putative mechanism(s) by which apoA-I may influence Aβ deposition is unknown. METHODS We evaluated the effect of apoA-I deletion on Aβ pathology, Aβ production and clearance from the brain in the Tg2576 mouse model of Alzheimer's disease (AD). RESULTS Contrary to previous reports, deletion of the APOA1 gene significantly reduced concentrations of insoluble Aβ40 and Aβ42 and reduced plaque load in both the parenchyma and blood vessels of apoA-I KO × Tg2576 mice compared to Tg2576 animals. This was not due to decreased Aβ production or alterations in Aβ species. Levels of soluble clusterin/apoJ were significantly higher in neurons of apoA-I KO mice compared to both wildtype (WT) and apoA-I KO × Tg2576 mice. In addition, clearance of Aβ along intramural periarterial drainage pathways was significantly higher in apoA-I KO mice compared to WT animals. CONCLUSION These data suggest that deletion of apoA-I is associated with increased clearance of Aβ and reduced parenchymal and vascular Aβ pathology in the Tg2576 model. These results suggest that peripheral dyslipidaemia can modulate the expression of apolipoproteins in the brain and may influence Aβ clearance and aggregation in AD.
Collapse
Affiliation(s)
- L Contu
- School of Life, Health and Chemical Sciences, STEM Faculty, The Open University, Milton Keynes, UK
| | - R O Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - C A Hawkes
- School of Life, Health and Chemical Sciences, STEM Faculty, The Open University, Milton Keynes, UK
| |
Collapse
|
41
|
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: 217] [Impact Index Per Article: 36.2] [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
|
42
|
Wiggs JL, Kang JH, Fan B, Levkovitch-Verbin H, Pasquale LR. A Role for Clusterin in Exfoliation Syndrome and Exfoliation Glaucoma? J Glaucoma 2018; 27 Suppl 1:S61-S66. [PMID: 29965900 PMCID: PMC8035929 DOI: 10.1097/ijg.0000000000000916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The multifunctional protein clusterin (CLU) is a secreted glycoprotein ubiquitously expressed throughout the body, including in the eye. Its primary function is to act as an extracellular molecular chaperone, preventing the precipitation and aggregation of misfolded extracellular proteins. Clusterin is commonly identified at fluid-tissue interfaces, and has been identified in most body fluids. It is a component of exfoliation material, and CLU mRNA is reduced in eyes with exfoliation syndrome compared with controls. SNPs located in the CLU genomic region have been associated with Alzheimer disease (AD) at the genome-wide level and several CLU SNPs located in an apparent regulatory region have been nominally associated with XFS/XFG in Caucasians with European ancestry and in south Indians. Interestingly, clusterin associates with altered elastic fibers in human photoaged skin and prevents UV-induced elastin aggregation in vitro. In light of the known geographic risk factors for XFS/XFG, which could include UV light, investigations of CLU-geographic interactions could be of interest. Future studies investigating rare CLU variation and other complex interactions including gene-gene interactions in XFS/XFG cases and controls may also be fruitful. Although CLU has been considered as a therapeutic target in AD, cancer and dry eye, a role for clusterin in XFS/XFG needs to be better defined before therapeutic approaches involving CLU can be entertained.
Collapse
Affiliation(s)
- Janey L. Wiggs
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Jae Hee Kang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - BaoJian Fan
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Hani Levkovitch-Verbin
- Goldschleger Eye Institute, Tel Hashomer, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Louis R. Pasquale
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Gulati T, Huang C, Caramia F, Raghu D, Paul PJ, Goode RJA, Keam SP, Williams SG, Haupt S, Kleifeld O, Schittenhelm RB, Gamell C, Haupt Y. Proteotranscriptomic Measurements of E6-Associated Protein (E6AP) Targets in DU145 Prostate Cancer Cells. Mol Cell Proteomics 2018; 17:1170-1183. [PMID: 29463595 DOI: 10.1074/mcp.ra117.000504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/18/2018] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer is a common cause of cancer-related death in men. E6AP (E6-Associated Protein), an E3 ubiquitin ligase and a transcription cofactor, is elevated in a subset of prostate cancer patients. Genetic manipulations of E6AP in prostate cancer cells expose a role of E6AP in promoting growth and survival of prostate cancer cells in vitro and in vivo However, the effect of E6AP on prostate cancer cells is broad and it cannot be explained fully by previously identified tumor suppressor targets of E6AP, promyelocytic leukemia protein and p27. To explore additional players that are regulated downstream of E6AP, we combined a transcriptomic and proteomic approach. We identified and quantified 16,130 transcripts and 7,209 proteins in castration resistant prostate cancer cell line, DU145. A total of 2,763 transcripts and 308 proteins were significantly altered on knockdown of E6AP. Pathway analyses supported the known phenotypic effects of E6AP knockdown in prostate cancer cells and in parallel exposed novel potential links of E6AP with cancer metabolism, DNA damage repair and immune response. Changes in expression of the top candidates were confirmed using real-time polymerase chain reaction. Of these, clusterin, a stress-induced chaperone protein, commonly deregulated in prostate cancer, was pursued further. Knockdown of E6AP resulted in increased clusterin transcript and protein levels in vitro and in vivo Concomitant knockdown of E6AP and clusterin supported the contribution of clusterin to the phenotype induced by E6AP. Overall, results from this study provide insight into the potential biological pathways controlled by E6AP in prostate cancer cells and identifies clusterin as a novel target of E6AP.
Collapse
Affiliation(s)
- Twishi Gulati
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Cheng Huang
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Franco Caramia
- §Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dinesh Raghu
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Piotr J Paul
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Robert J A Goode
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Simon P Keam
- §Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Scott G Williams
- ‖Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sue Haupt
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Oded Kleifeld
- **Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ralf B Schittenhelm
- ¶Monash Biomedical Proteomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Cristina Gamell
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ygal Haupt
- From the ‡The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; .,§Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,‡‡Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,§§Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia.,¶¶Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
44
|
Rohne P, Wolf S, Dörr C, Ringen J, Holtz A, Gollan R, Renner B, Prochnow H, Baiersdörfer M, Koch-Brandt C. Exposure of vital cells to necrotic cell lysates induce the IRE1α branch of the unfolded protein response and cell proliferation. Cell Stress Chaperones 2018; 23:77-88. [PMID: 28687980 PMCID: PMC5741583 DOI: 10.1007/s12192-017-0825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/15/2017] [Accepted: 06/16/2017] [Indexed: 10/19/2022] Open
Abstract
Necrosis is a form of cell death that is detrimental to the affected tissue because the cell ruptures and releases its content (reactive oxygen species among others) into the extracellular space. Clusterin (CLU), a cytoprotective extracellular chaperone has been shown to be upregulated in the face of necrosis. We here show that in addition to CLU upregulation, necrotic cell lysates induce JNK/SAPK signaling, the IRE1α branch of the unfolded protein response (UPR), the MAPK/ERK1/2, and the mTOR signaling pathways and results in an enhanced proliferation of the vital surrounding cells. We name this novel response mechanism: Necrosis-induced Proliferation (NiP).
Collapse
Affiliation(s)
- Philipp Rohne
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Steven Wolf
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Pathology, The University of Chicago, Chicago, IL USA
| | - Carolin Dörr
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Julia Ringen
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Andrew Holtz
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - René Gollan
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Benjamin Renner
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Hans Prochnow
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Braunschweig, Germany
| | - Markus Baiersdörfer
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Claudia Koch-Brandt
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| |
Collapse
|
45
|
Habiel DM, Camelo A, Espindola M, Burwell T, Hanna R, Miranda E, Carruthers A, Bell M, Coelho AL, Liu H, Pilataxi F, Clarke L, Grant E, Lewis A, Moore B, Knight DA, Hogaboam CM, Murray LA. Divergent roles for Clusterin in Lung Injury and Repair. Sci Rep 2017; 7:15444. [PMID: 29133960 PMCID: PMC5684342 DOI: 10.1038/s41598-017-15670-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023] Open
Abstract
Lung fibrosis is an unabated wound healing response characterized by the loss and aberrant function of lung epithelial cells. Herein, we report that extracellular Clusterin promoted epithelial cell apoptosis whereas intracellular Clusterin maintained epithelium viability during lung repair. Unlike normal and COPD lungs, IPF lungs were characterized by significantly increased extracellular Clusterin whereas the inverse was evident for intracellular Clusterin. In vitro and in vivo studies demonstrated that extracellular Clusterin promoted epithelial cell apoptosis while intercellular Clusterin modulated the expression of the DNA repair proteins, MSH2, MSH6, OGG1 and BRCA1. The fibrotic response in Clusterin deficient (CLU-/-) mice persisted after bleomycin and it was associated with increased DNA damage, reduced DNA repair responses, and elevated cellular senescence. Remarkably, this pattern mirrored that observed in IPF lung tissues. Together, our results show that cellular localization of Clusterin leads to divergent effects on epithelial cell regeneration and lung repair during fibrosis.
Collapse
Affiliation(s)
- David M Habiel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Ana Camelo
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Cambridge, United Kingdom
| | - Milena Espindola
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy Burwell
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, USA
| | - Richard Hanna
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD, USA
| | - Elena Miranda
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, USA
| | - Alan Carruthers
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Cambridge, United Kingdom
| | - Matthew Bell
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Cambridge, United Kingdom
| | - Ana Lucia Coelho
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hao Liu
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, USA
| | | | - Lori Clarke
- Molecular Biology, MedImmune LLC, Gaithersburg, MD, USA
| | - Ethan Grant
- Translational Medicine, MedImmune LLC, Gaithersburg, MD, USA
| | - Arthur Lewis
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Cambridge, United Kingdom
| | - Bethany Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Cory M Hogaboam
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lynne A Murray
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Cambridge, United Kingdom.
| |
Collapse
|
46
|
Gregory JM, Whiten DR, Brown RA, Barros TP, Kumita JR, Yerbury JJ, Satapathy S, McDade K, Smith C, Luheshi LM, Dobson CM, Wilson MR. Clusterin protects neurons against intracellular proteotoxicity. Acta Neuropathol Commun 2017; 5:81. [PMID: 29115989 PMCID: PMC5678579 DOI: 10.1186/s40478-017-0481-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022] Open
Abstract
It is now widely accepted in the field that the normally secreted chaperone clusterin is redirected to the cytosol during endoplasmic reticulum (ER) stress, although the physiological function(s) of this physical relocation remain unknown. We have examined in this study whether or not increased expression of clusterin is able to protect neuronal cells against intracellular protein aggregation and cytotoxicity, characteristics that are strongly implicated in a range of neurodegenerative diseases. We used the amyotrophic lateral sclerosis-associated protein TDP-43 as a primary model to investigate the effects of clusterin on protein aggregation and neurotoxicity in complementary in vitro, neuronal cell and Drosophila systems. We have shown that clusterin directly interacts with TDP-43 in vitro and potently inhibits its aggregation, and observed that in ER stressed neuronal cells, clusterin co-localized with TDP-43 and specifically reduced the numbers of cytoplasmic inclusions. We further showed that the expression of TDP-43 in transgenic Drosophila neurons induced ER stress and that co-expression of clusterin resulted in a dramatic clearance of mislocalized TDP-43 from motor neuron axons, partially rescued locomotor activity and significantly extended lifespan. We also showed that in Drosophila photoreceptor cells, clusterin co-expression gave ER stress-dependent protection against proteotoxicity arising from both Huntingtin-Q128 and mutant (R406W) human tau. We therefore conclude that increased expression of clusterin can provide an important defense against intracellular proteotoxicity under conditions that mimic specific features of neurodegenerative disease.
Collapse
Affiliation(s)
- Jenna M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, 49 Little France Crescent-Chancellor, Edinburgh, EH16 4SB, UK
| | - Daniel R Whiten
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Rebecca A Brown
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Teresa P Barros
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Janet R Kumita
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Sandeep Satapathy
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Karina McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Leila M Luheshi
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
47
|
Li X, Song Y, Sanders CR, Buxbaum JN. Transthyretin Suppresses Amyloid-β Secretion by Interfering with Processing of the Amyloid-β Protein Precursor. J Alzheimers Dis 2017; 52:1263-75. [PMID: 27079720 DOI: 10.3233/jad-160033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In Alzheimer's disease (AD), most hippocampal and cortical neurons show increased staining with anti-transthyretin (TTR) antibodies. Genetically programmed overexpression of wild type human TTR suppressed the neuropathologic and behavioral abnormalities in APP23 AD model mice and TTR-Aβ complexes have been isolated from some human AD brains and those of APP23 transgenic mice. In the present study, in vitro NMR analysis showed interaction between the hydrophobic thyroxine binding pocket of TTR and the cytoplasmic loop of the C99 fragment released by β-secretase cleavage of AβPP, with Kd = 86±9 μM. In cultured cells expressing both proteins, the interaction reduced phosphorylation of C99 (at T668) and suppressed its cleavage by γ-secretase, significantly decreasing Aβ secretion. Coupled with its previously demonstrated capacity to inhibit Aβ aggregation (with the resultant cytotoxicity in tissue culture) and its regulation by HSF1, these findings indicate that TTR can behave as a stress responsive multimodal suppressor of AD pathogenesis.
Collapse
Affiliation(s)
- Xinyi Li
- Janssen Research & Development, LLC, Johnson & Johnson, San Diego, CA, USA.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Yuanli Song
- Bristol-Myers Squibb, Biologics Process Development, Devens, MA, USA.,Department of Biochemistry and Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Charles R Sanders
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joel N Buxbaum
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
48
|
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: 15] [Impact Index Per Article: 1.9] [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
|
49
|
Wang Y, Qin X, Paudel HK. Amyloid-β breaks brain's neuroprotection. Oncotarget 2017; 8:48533. [PMID: 28562319 PMCID: PMC5564705 DOI: 10.18632/oncotarget.18246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Yunling Wang
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill university, Montreal, Quebec, Canada
| | - Xike Qin
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill university, Montreal, Quebec, Canada
| | - Hemant K Paudel
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill university, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Plate L, Wiseman RL. Regulating Secretory Proteostasis through the Unfolded Protein Response: From Function to Therapy. Trends Cell Biol 2017. [PMID: 28647092 DOI: 10.1016/j.tcb.2017.05.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Imbalances in secretory proteostasis induced by genetic, environmental, or aging-related insults are pathologically associated with etiologically diverse protein misfolding diseases. To protect the secretory proteome from these insults, organisms evolved stress-responsive signaling pathways that regulate the composition and activity of biologic pathways involved in secretory proteostasis maintenance. The most prominent of these is the endoplasmic reticulum (ER) unfolded protein response (UPR), which functions to regulate ER proteostasis in response to ER stress. While the signaling mechanisms involved in UPR activation are well defined, the impact of UPR activation on secretory proteostasis is only now becoming clear. Here, we highlight recent reports defining how activation of select UPR signaling pathways influences proteostasis within the ER and downstream secretory environments. Furthermore, we describe recent evidence that highlights the therapeutic potential for targeting UPR signaling pathways to correct pathologic disruption in secretory proteostasis associated with diverse types of protein misfolding diseases.
Collapse
Affiliation(s)
- Lars Plate
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|