1
|
Turkieh A, Weber L, Chwastyniak M, Baydar S, Beseme O, Ernst M, Ye Q, Amouyel P, Podesser BK, Kiss A, Pinet F. Regulation of Clusterin in the Heart and Plasma of Mice After Transverse Aortic Constriction. J Cell Mol Med 2024; 28:e70290. [PMID: 39671261 PMCID: PMC11640023 DOI: 10.1111/jcmm.70290] [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: 04/22/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024] Open
Abstract
Chronic pressure overload induces adverse cardiac remodelling characterised by left ventricular (LV) hypertrophy and fibrosis, leading to heart failure (HF). Identification of new biomarkers for adverse cardiac remodelling enables us to better understand this process and, consequently, to prevent HF. We recently identified clusterin (CLU) as a biomarker of cardiac remodelling and HF after myocardial infarction. The aim of this study was to investigate whether CLU expression is regulated in the heart and could be used as an indicator of adverse cardiac remodelling in response to pressure overload. We quantified CLU in the LV of mice subjected to transverse aortic constriction (TAC) and observed increased CLU mRNA levels and its mature protein form (m-CLU) compared to the sham. Interestingly, CLU mRNA levels were positively correlated with pro-hypertrophic (ANP, BNP, B-MHC), pro- and anti-fibrotic (TGFb, ColI and CILP) genes. In addition, m-CLU was positively correlated with LV hypertrophy, LV end diastolic and systolic diameters, and negatively correlated with LV ejection fraction. Finally, we observed that m-CLU levels only increased in TAC mice with severe cardiac remodelling and dysfunction without any significant difference in plasma CLU levels. This is the first study to demonstrate that cardiac expression of CLU is induced in the LV of TAC mice during adverse cardiac remodelling. However, plasma CLU levels could not be used as biomarkers of TAC-induced cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Annie Turkieh
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGEUniversité de LilleLilleFrance
| | - Lukas Weber
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Maggy Chwastyniak
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGEUniversité de LilleLilleFrance
| | - Simge Baydar
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Olivia Beseme
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGEUniversité de LilleLilleFrance
| | - Matthias Ernst
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Quianling Ye
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Philippe Amouyel
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGEUniversité de LilleLilleFrance
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational SurgeryMedical University of ViennaViennaAustria
| | - Florence Pinet
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGEUniversité de LilleLilleFrance
| |
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
|
Puig N, Rives J, Gil-Millan P, Miñambres I, Ginel A, Tauron M, Bonaterra-Pastra A, Hernández-Guillamon M, Pérez A, Sánchez-Quesada JL, Benitez S. Apolipoprotein J protects cardiomyocytes from lipid-mediated inflammation and cytotoxicity induced by the epicardial adipose tissue of diabetic patients. Biomed Pharmacother 2024; 175:116779. [PMID: 38776681 DOI: 10.1016/j.biopha.2024.116779] [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: 03/06/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Diabetic patients present increased volume and functional alterations in epicardial adipose tissue (EAT). We aimed to analyze EAT from type 2 diabetic patients and the inflammatory and cytotoxic effects induced on cardiomyocytes. Furthermore, we analyzed the cardioprotective role of apolipoprotein J (apoJ). EAT explants were obtained from nondiabetic patients (ND), diabetic patients without coronary disease (DM), and DM patients with coronary disease (DM-C) after heart surgery. Morphological characteristics and gene expression were evaluated. Explants were cultured for 24 h and the content of nonesterified fatty acids (NEFA) and sphingolipid species in secretomes was evaluated by lipidomic analysis. Afterwards, secretomes were added to AC16 human cardiomyocytes for 24 h in the presence or absence of cardioprotective molecules (apoJ and HDL). Cytokine release and apoptosis/necrosis were assessed by ELISA and flow cytometry. The EAT from the diabetic samples showed altered expression of genes related to lipid accumulation, insulin resistance, and inflammation. The secretomes from the DM samples presented an increased ratio of pro/antiatherogenic ceramide (Cer) species, while those from DM-C contained the highest concentration of saturated NEFA. DM and DM-C secretomes promoted inflammation and cytotoxicity on AC16 cardiomyocytes. Exogenous Cer16:0, Cer24:1, and palmitic acid reproduced deleterious effects in AC16 cells. These effects were attenuated by exogenous apoJ. Diabetic secretomes promoted inflammation and cytotoxicity in cardiomyocytes. This effect was exacerbated in the secretomes of the DM-C samples. The increased content of specific NEFA and ceramide species seems to play a key role in inducing such deleterious effects, which are attenuated by apoJ.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - José Rives
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Pedro Gil-Millan
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Inka Miñambres
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Antonino Ginel
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Manel Tauron
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio Pérez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - José Luís Sánchez-Quesada
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Sonia Benitez
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| |
Collapse
|
4
|
Fiet MD, Wu L, Koo A, Woudstra L, Niessen HWM, Krijnen PAJ. Clusterin is increased in morphologically normal cardiomyocytes in lymphocytic myocarditis. Eur J Clin Invest 2023; 53:e13905. [PMID: 36349473 DOI: 10.1111/eci.13905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Mitchell D Fiet
- Department of Pathology, Amsterdam University Medical Center, Location VUmc and AMC, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Linghe Wu
- Department of Pathology, Amsterdam University Medical Center, Location VUmc and AMC, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Amy Koo
- Department of Pathology, Amsterdam University Medical Center, Location VUmc and AMC, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Linde Woudstra
- Department of Pathology, Amsterdam University Medical Center, Location VUmc and AMC, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Hans W M Niessen
- Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands.,Department of Cardiac Surgery, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam University Medical Center, Location VUmc and AMC, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Impaired Extracellular Proteostasis in Patients with Heart Failure. Arch Med Res 2023; 54:211-222. [PMID: 36797157 DOI: 10.1016/j.arcmed.2023.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Proteostasis impairment and the consequent increase of amyloid burden in the myocardium have been associated with heart failure (HF) development and poor prognosis. A better knowledge of the protein aggregation process in biofluids could assist the development and monitoring of tailored interventions. AIM To compare the proteostasis status and protein's secondary structures in plasma samples of patients with HF with preserved ejection fraction (HFpEF), patients with HF with reduced ejection fraction (HFrEF), and age-matched individuals. METHODS A total of 42 participants were enrolled in 3 groups: 14 patients with HFpEF, 14 patients with HFrEF, and 14 age-matched individuals. Proteostasis-related markers were analyzed by immunoblotting techniques. Fourier Transform Infrared (FTIR) Spectroscopy in Attenuated Total Reflectance (ATR) was applied to assess changes in the protein's conformational profile. RESULTS Patients with HFrEF showed an elevated concentration of oligomeric proteic species and reduced clusterin levels. ATR-FTIR spectroscopy coupled with multivariate analysis allowed the discrimination of HF patients from age-matched individuals in the protein amide I absorption region (1700-1600 cm-1), reflecting changes in protein conformation, with a sensitivity of 73 and a specificity of 81%. Further analysis of FTIR spectra showed significantly reduced random coils levels in both HF phenotypes. Also, compared to the age-matched group, the levels of structures related to fibril formation were significantly increased in patients with HFrEF, whereas the β-turns were significantly increased in patients with HFpEF. CONCLUSION Both HF phenotypes showed a compromised extracellular proteostasis and different protein conformational changes, suggesting a less efficient protein quality control system.
Collapse
|
6
|
The Emerging Roles of Extracellular Chaperones in Complement Regulation. Cells 2022; 11:cells11233907. [PMID: 36497163 PMCID: PMC9738919 DOI: 10.3390/cells11233907] [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: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
The immune system is essential to protect organisms from internal and external threats. The rapidly acting, non-specific innate immune system includes complement, which initiates an inflammatory cascade and can form pores in the membranes of target cells to induce cell lysis. Regulation of protein homeostasis (proteostasis) is essential for normal cellular and organismal function, and has been implicated in processes controlling immunity and infection. Chaperones are key players in maintaining proteostasis in both the intra- and extracellular environments. Whilst intracellular proteostasis is well-characterised, the role of constitutively secreted extracellular chaperones (ECs) is less well understood. ECs may interact with invading pathogens, and elements of the subsequent immune response, including the complement pathway. Both ECs and complement can influence the progression of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis, as well as other diseases including kidney diseases and diabetes. This review will examine known and recently discovered ECs, and their roles in immunity, with a specific focus on the complement pathway.
Collapse
|
7
|
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
|
8
|
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
|
9
|
Kang M, Seong Y, Mahmud J, Nguyen BT. Obscurin and Clusterin Elevation in Serum of Acute Myocardial Infarction Patients. B KOREAN CHEM SOC 2020. [DOI: 10.1002/bkcs.11955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Min‐Jung Kang
- Molecular Recognition Research CenterKorea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology Seoul 02792 Republic of Korea
| | - Yunseo Seong
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology Seoul 02792 Republic of Korea
| | - Joyeta Mahmud
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology Seoul 02792 Republic of Korea
| | - Binh Thanh Nguyen
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology Seoul 02792 Republic of Korea
| |
Collapse
|
10
|
Turkieh A, Porouchani S, Beseme O, Chwastyniak M, Amouyel P, Lamblin N, Balligand JL, Bauters C, Pinet F. Increased clusterin levels after myocardial infarction is due to a defect in protein degradation systems activity. Cell Death Dis 2019; 10:608. [PMID: 31406108 PMCID: PMC6691115 DOI: 10.1038/s41419-019-1857-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/22/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022]
Abstract
Clusterin (CLU) is induced in many organs after tissue injury or remodeling. Recently, we show that CLU levels are increased in plasma and left ventricle (LV) after MI, however, the mechanisms involved are not yet elucidated. On the other hand, it has been shown that the activity of the protein degradation systems (PDS) is affected after MI with a decrease in ubiquitin proteasome system (UPS) and an increase in macroautophagy. The aim of this study was to decipher if the increased CLU levels after MI are in part due to the alteration of PDS activity. Rat neonate cardiomyocytes (NCM) were treated with different modulators of UPS and macroautophagy in order to decipher their role in CLU expression, secretion, and degradation. We observed that inhibition of UPS activity in NCM increased CLU mRNA levels, its intracellular protein levels (p-CLU and m-CLU) and its secreted form (s-CLU). Macroautophagy was also induced after MG132 treatment but is not active. The inhibition of macroautophagy induction in MG132-treated NCM increased CLU mRNA and m-CLU levels, but not s-CLU compared to NCM only treated by MG132. We also demonstrate that CLU can be degraded in NCM through proteasome and lysosome by a macroautophagy independent pathway. In another hand, CLU silencing in NCM has no effect either on macroautophagy or apoptosis induced by MG132. However, the overexpression of CLU secreted isoform in H9c2 cells, but not in NCM decreased apoptosis after MG132 treatment. Finally, we observed that increased CLU levels in hypertrophied NCM and in failing human hearts are associated with proteasome inhibition and macroautophagy alteration. All these data suggest that increased CLU expression and secretion after MI is, in part, due to a defect of UPS and macroautophagy activities in the heart and may have a protective effect by decreasing apoptosis induced by proteasome inhibition.
Collapse
Affiliation(s)
- Annie Turkieh
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Sina Porouchani
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Olivia Beseme
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Maggy Chwastyniak
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Philippe Amouyel
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France
| | - Nicolas Lamblin
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Jean-Luc Balligand
- Institut de Recherche Experimentale et Clinique, Pole of Pharmacology and Therapeutics and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Bauters
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France.,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France
| | - Florence Pinet
- Inserm, University of Lille, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000, Lille, France. .,Fédération Hospitalière Universitaire (FHU), REMOD-VHF, Lille, France.
| |
Collapse
|
11
|
Ma Y, Gong Z, Nan K, Qi S, Chen Y, Ding C, Wang D, Ru L. Apolipoprotein-J blocks increased cell injury elicited by ox-LDL via inhibiting ROS-CaMKII pathway. Lipids Health Dis 2019; 18:117. [PMID: 31113434 PMCID: PMC6530009 DOI: 10.1186/s12944-019-1066-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
Background Oxidized low-density lipoprotein (ox-LDL) is crucial in cardiac injury. Apolipoprotein-J (ApoJ) contributes to antiapoptotic effects in the heart. We aimed to evaluate the protective effects of ApoJ against ox-LDL cytotoxicity in Neonatal rat ventricular cells (NRVCs). Methods and results NRVCs were damaged by exposure to ox-LDL, as shown by increased caspase-3/7 activity, enhanced caspase-3 expression, and decreased cell viability. ApoJ overexpression, using an adenovirus vector, significantly reduced ox-LDL-induced cell injury. ApoJ also prevented ox-LDL from augmenting reactive oxygen species (ROS) production, as demonstrated by elevated Nox2/gp91phox and P47 expression. Furthermore, ApoJ overexpression reduced CaMKIIδ expression elicited by ox-LDL in cultured NRVCs. Upregulating CaMKIIδ activity, mediated by ox-LDL, was significantly inhibited by ApoJ overexpression. A CaMKIIδ inhibitor, KN93, prevented ApoJ’s protective effect against ox-LDL cytotoxicity. A ROS scavenger, Mn (III)meso-tetrakis (4-benzoic acid) porphyrin (Mn (III)TBAP), also attenuated CaMKIIδ’s increased expression and activity, induced by ox-LDL, and showed similar results to ApoJ by attenuating ox-LDL-induced cell damage, as ApoJ did. Conclusions ApoJ confers cytoprotection to NRVCs against ox-LDL cytotoxicity through the ROS-CaMKII pathways.
Collapse
Affiliation(s)
- Yanzhuo Ma
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Zhi Gong
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Kai Nan
- Health and Medical Development Research Center of Hebei Province, Shijiazhuang, Hebei, China
| | - Shuying Qi
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Yu Chen
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Chao Ding
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Dongmei Wang
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China
| | - Leisheng Ru
- Department of Cardiology, Bethune International Peace Hospital, 398, Zhongshan Road, Shijiazhuang, 050082, Hebei, China.
| |
Collapse
|
12
|
Zhu H, Liu M, Zhai T, Pan H, Wang L, Yang H, Yan K, Gong F, Zeng Y. High serum clusterin levels are associated with premature coronary artery disease in a Chinese population. Diabetes Metab Res Rev 2019; 35:e3128. [PMID: 30659732 DOI: 10.1002/dmrr.3128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND Clusterin plays an important role in the cardiovascular system, and serum levels of clusterin are higher in coronary artery disease patients. Here, we measured serum clusterin levels in premature coronary artery disease (PCAD) patients and explored the association of these levels with PCAD risk. METHODS Serum samples and general clinical information were obtained from 672 subjects including 364 PCAD subjects, 126 non-PCAD subjects, and 182 controls. RESULTS Serum clusterin levels were higher in PCAD patients than in controls, particularly in males with body mass index (BMI) < 25 kg/m2 (P < 0.0001). Compared with the lowest tertile of clusterin, the odds ratio of PCAD in the highest tertile was higher in both a univariate and three adjustment models, and it was 3.146-fold higher in Model 3. This association was especially significant in subgroups with BMI < 25 kg/m2 , total cholesterol < 5.7 mmol/L, high-density lipoprotein cholesterol ≥ 1.0 mmol/L, Urea < 7.14 mmol/L, and estimated glomerular filtration rate < 90 mL/min/1.73 m2 . Serum clusterin may be a potential diagnostic biomarker for PCAD (sensitivity 60.7%, specificity 51.6%, area under the curve 0.595 [95% CI, 0.544-0.647], P < 0.0001), and a combination of clusterin with clinical variables in Model 3 resulted in improved diagnostic accuracy (sensitivity 86.3%, specificity 64.2%, area under the curve 0.829 [95% CI, 0.782-0.877], P < 0.0001). CONCLUSIONS Serum clusterin levels were increased in PCAD patients, especially for males with BMI < 25 kg/m2 . Higher clusterin levels were independently associated with the presence of PCAD, particularly in subjects with normal BMI, lower total cholesterol, urea, estimated glomerular filtration rate, and higher high-density lipoprotein cholesterol. Clusterin might be a potential diagnostic biomarker for PCAD patients, especially in combination with clinical variables.
Collapse
Affiliation(s)
- Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Meijuan Liu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tianshu Zhai
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Kemin Yan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yong Zeng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Pereira RM, Mekary RA, da Cruz Rodrigues KC, Anaruma CP, Ropelle ER, da Silva ASR, Cintra DE, Pauli JR, de Moura LP. Protective molecular mechanisms of clusterin against apoptosis in cardiomyocytes. Heart Fail Rev 2019; 23:123-129. [PMID: 28948410 DOI: 10.1007/s10741-017-9654-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Loss of cardiomyocytes occurs with aging and contributes to cardiovascular complications. In the present study, we highlighted the role of clusterin, a protein that has recently been associated with the protection of cardiomyocytes from apoptosis. Clusterin protects cardiac cells against damage from myocardial infarction, transplant, or myocarditis. Clusterin can act directly or indirectly on apoptosis by regulating several intracellular pathways. These pathways include (1) the oxidant and inflammatory program, (2) insulin growth factor 1 (IGF-1) pathway, (3) KU70 / BCL-2-associated X protein (BAX) pathway, (4) tumor necrosis factor alpha (TNF-α) pathway, (5) BCL-2 antagonist of cell death (BAD) pathway, and (6) mitogen-activated protein kinase (MAPK) pathway. Given the key role of clusterin in preventing loss of cardiac tissue, modulating the expression and function of this protein carries the potential of improving cardiovascular care in the future.
Collapse
Affiliation(s)
- Rodrigo Martins Pereira
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rania A Mekary
- Department of Pharmaceutical Business and Administrative Sciences, MCPHS University, Boston, MA, USA.,Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Cristina da Cruz Rodrigues
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Chadi Pellegrini Anaruma
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science, University of Campinas, 1300 Pedro Zaccaria St, Limeira, São Paulo, Brazil. .,CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| |
Collapse
|
14
|
p47phox-Dependent Reactive Oxygen Species Stimulate Nuclear Translocation of the FoxO1 Transcription Factor During Metabolic Inhibition in Cardiomyoblasts. Cell Biochem Biophys 2018; 76:401-410. [PMID: 29956081 PMCID: PMC6097050 DOI: 10.1007/s12013-018-0847-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/11/2018] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) control forkhead box O (FOXO) transcription factor activity by influencing their nuclear translocation. However, knowledge of the ROS cellular source(s) involved herein remains scarce. Recently, we have shown p47phox-dependent activation of ROS-producing NADPH oxidase (NOX) at the nuclear pore in H9c2 rat cardiomyoblasts in response to ischemia. This localizes NOX perfectly to affect protein nuclear translocation, including that of transcription factors. In the current study, involvement of p47phox-dependent production of ROS in the nuclear translocation of FOXO1 was analyzed in H9c2 cells following 4 h of metabolic inhibition (MI), which mimics the effects of ischemia. Nuclear translocation of FOXO1 was determined by quantitative digital-imaging fluorescence and western blot analysis. Subsequently, the effect of inhibiting p47phox-dependent ROS production by short hairpin RNA (shRNA) transfection on FOXO1 translocation was analyzed by digital-imaging microscopy. MI induced a significant translocation of FOXO1 into the nucleus. Transfection with p47phox-shRNA successfully knocked-down p47phox expression, reduced nuclear nitrotyrosine production, an indirect marker for ROS production, and inhibited the nuclear translocation of FOXO1 following MI. With these results, we show for the first time that nuclear import of FOXO1 induced by MI in H9c2 depends critically on p47phox-mediated ROS production.
Collapse
|
15
|
Turkieh A, Fertin M, Bouvet M, Mulder P, Drobecq H, Lemesle G, Lamblin N, de Groote P, Porouchani S, Chwastyniak M, Beseme O, Amouyel P, Mouquet F, Balligand JL, Richard V, Bauters C, Pinet F. Expression and Implication of Clusterin in Left Ventricular Remodeling After Myocardial Infarction. Circ Heart Fail 2018; 11:e004838. [DOI: 10.1161/circheartfailure.117.004838] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/16/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Annie Turkieh
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Marie Fertin
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Marion Bouvet
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Paul Mulder
- Institut Pasteur de Lille, Université de Lille, France. Inserm U1096, FHU-REMOD-VHF, Normandie University, University of Rouen, France (P.M., V.R.)
| | - Hervé Drobecq
- UMR 8161-M3T-Mechanisms of Tumorigenesis and Target Therapies, CNRS (H.D.)
| | - Gilles Lemesle
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Nicolas Lamblin
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Pascal de Groote
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
| | - Sina Porouchani
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Maggy Chwastyniak
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Olivia Beseme
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| | - Philippe Amouyel
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- CHU Lille, Service de Santé Publique, Épidémiologie, Économie de la Santé et Prévention, France (P.A.)
| | - Frédéric Mouquet
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
| | - Jean-Luc Balligand
- Institut de Recherche Experimentale et Clinique, Pole of Pharmacology and Therapeutics and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium (J.-L.B.)
| | - Vincent Richard
- Institut Pasteur de Lille, Université de Lille, France. Inserm U1096, FHU-REMOD-VHF, Normandie University, University of Rouen, France (P.M., V.R.)
| | - Christophe Bauters
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, France (G.L., N.L., P.d.G., F.M., C.B.)
- Faculté de Médecine de l’Université de Lille, France (G.L., N.L., P.A., C.B.)
- FACT, French Alliance for Cardiovascular Trials, Paris, France (G.L., N.L., C.B.)
| | - Florence Pinet
- INSERM U1167-RID-AGE, CHU Lille, FHU-REMOD-VHF (A.T., M.F., M.B., N.L., P.d.G., S.P., M.C., O.B., P.A., C.B., F.P.)
| |
Collapse
|
16
|
On the value of therapeutic interventions targeting the complement system in acute myocardial infarction. Transl Res 2017; 182:103-122. [PMID: 27810412 DOI: 10.1016/j.trsl.2016.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/12/2023]
Abstract
The complement system plays an important role in the inflammatory response subsequent to acute myocardial infarction (AMI). The aim of this study is to create a systematic overview of studies that have investigated therapeutic administration of complement inhibitors in both AMI animal models and human clinical trials. To enable extrapolation of observations from included animal studies toward post-AMI clinical trials, ex vivo studies on isolated hearts and proof-of-principle studies on inhibitor administration before experimental AMI induction were excluded. Positive therapeutic effects in AMI animal models have been described for cobra venom factor, soluble complement receptor 1, C1-esterase inhibitor (C1-inh), FUT-175, C1s-inhibitor, anti-C5, ADC-1004, clusterin, and glycosaminoglycans. Two types of complement inhibitors have been tested in clinical trials, being C1-inh and anti-C5. Pexelizumab (anti-C5) did not result in reproducible beneficial effects for AMI patients. Beneficial effects were reported in AMI patients for C1-inhibitor, albeit in small patient groups. In general, despite the absence of consistent positive effects in clinical trials thus far, the complement system remains a potentially interesting target for therapy in AMI patients. Based on the study designs of previous animal studies and clinical trials, we discuss several issues which require attention in the design of future studies: adjustment of clinical trial design to precise mechanism of action of administered inhibitor, optimizing the duration of therapy, and optimization of time point(s) on which therapeutic effects will be evaluated.
Collapse
|
17
|
Clusterin/apolipoprotein J is independently associated with survival in patients with chronic heart failure. J Clin Lipidol 2017; 11:178-184. [DOI: 10.1016/j.jacl.2016.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/26/2016] [Indexed: 12/20/2022]
|
18
|
Begieneman MPV, Ter Horst EN, Rijvers L, Meinster E, Leen R, Pankras JE, Fritz J, Kubat B, Musters RJP, van Kuilenburg ABP, Stap J, Niessen HWM, Krijnen PAJ. Dopamine induces lipid accumulation, NADPH oxidase-related oxidative stress, and a proinflammatory status of the plasma membrane in H9c2 cells. Am J Physiol Heart Circ Physiol 2016; 311:H1097-H1107. [PMID: 27521422 DOI: 10.1152/ajpheart.00633.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 07/15/2016] [Indexed: 01/08/2023]
Abstract
Excess catecholamine levels are suggested to be cardiotoxic and to underlie stress-induced heart failure. The cardiotoxic effects of norepinephrine and epinephrine are well recognized. However, although cardiac and circulating dopamine levels are also increased in stress cardiomyopathy patients, knowledge regarding putative toxic effects of excess dopamine levels on cardiomyocytes is scarce. We now studied the effects of elevated dopamine levels in H9c2 cardiomyoblasts. H9c2 cells were cultured and treated with dopamine (200 μM) for 6, 24, and 48 h. Subsequently, the effects on lipid accumulation, cell viability, flippase activity, reactive oxygen species (ROS) production, subcellular NADPH oxidase (NOX) protein expression, and ATP/ADP and GTP/GDP levels were analyzed. Dopamine did not result in cytotoxic effects after 6 h. However, after 24 and 48 h dopamine treatment induced a significant increase in lipid accumulation, nitrotyrosine levels, indicative of ROS production, and cell death. In addition, dopamine significantly reduced flippase activity and ATP/GTP levels, coinciding with phosphatidylserine exposure on the outer plasma membrane. Furthermore, dopamine induced a transient increase in cytoplasmic and (peri)nucleus NOX1 and NOX4 expression after 24 h that subsided after 48 h. Moreover, while dopamine induced a similar transient increase in cytoplasmic NOX2 and p47phox expression, in the (peri)nucleus this increased expression persisted for 48 h where it colocalized with ROS. Exposure of H9c2 cells to elevated dopamine levels induced lipid accumulation, oxidative stress, and a proinflammatory status of the plasma membrane. This can, in part, explain the inflammatory response in patients with stress-induced heart failure.
Collapse
Affiliation(s)
- Mark P V Begieneman
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands; .,Netherlands Forensic Institute, The Hague, the Netherlands.,Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Ellis N Ter Horst
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands.,Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - Liza Rijvers
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Elisa Meinster
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - René Leen
- Laboratory Genetic Metabolic Diseases and Department of Pediatrics/Emma's Children Hospital, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Jeannette E Pankras
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jan Fritz
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Bela Kubat
- Netherlands Forensic Institute, The Hague, the Netherlands.,Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - René J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Jan Stap
- Core Facility Cellular Imaging/LCAM-AMC, Amsterdam, the Netherlands; and
| | - Hans W M Niessen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands.,Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, the Netherlands.,Department of Cardiothoracic Surgery, VU University Medical Center, Amsterdam, the Netherlands
| | - Paul A J Krijnen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands.,Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Yu B, Yang Y, Liu H, Gong M, Millard RW, Wang YG, Ashraf M, Xu M. Clusterin/Akt Up-Regulation Is Critical for GATA-4 Mediated Cytoprotection of Mesenchymal Stem Cells against Ischemia Injury. PLoS One 2016; 11:e0151542. [PMID: 26962868 PMCID: PMC4786134 DOI: 10.1371/journal.pone.0151542] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/29/2016] [Indexed: 01/12/2023] Open
Abstract
Background Clusterin (Clu) is a stress-responding protein with multiple biological functions. Our preliminary microarray studies show that clusterin was prominently upregulated in mesenchymal stem cells (MSCs) overexpressing GATA-4 (MSCGATA-4). We hypothesized that the upregulation of clusterin is involved in overexpression of GATA-4 mediated cytoprotection. Methods MSCs harvested from bone marrow of rats were transduced with GATA-4. The expression of clusterin in MSCs was further confirmed by real-time PCR and western blotting. Simulation of ischemia was achieved by exposure of MSCs to a hypoxic environment. Lactate dehydrogenase (LDH) released from MSCs was served as a biomarker of cell injury and MTs uptake was used to estimate cell viability. Mitochondrial function was evaluated by measuring mitochondrial membrane potential (ΔΨm) and caspase 3/7 activity. Results (1) Clusterin expression was up-regulated in MSCGATA-4 compared to control MSCs transfected with empty-vector (MSCNull). MSCGATA-4 were tolerant to 72 h hypoxia exposure as shown by reduced LDH release and higher MTs uptake. This protection was abrogated by transfecting Clu-siRNA into MSCGATA-4. (2) Exogenous clusterin significantly decreased LDH release and increased MSC survival in hypoxic environment. Moreover, ΔΨm was maintained and caspase 3/7 activity was reduced by clusterin in a concentration-dependent manner. (3) p-Akt expression in MSCs was upregulated following pre-treatment with clusterin, with no change in total Akt. Moreover, cytoprotection mediated by clusterin was partially abrogated by Akt inhibitor LY294002. Conclusions Clusterin/Akt signaling pathway is involved in GATA-4 mediated cytoprotection against hypoxia stress. It is suggested that clusterin may be therapeutically exploited in MSC based therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Bin Yu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Yueting Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Huan Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Min Gong
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Ronald W. Millard
- Department of Pharmacology & Cell Biophysics, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Yi-Gang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Muhammad Ashraf
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
20
|
Endogenous C1-inhibitor production and expression in the heart after acute myocardial infarction. Cardiovasc Pathol 2015; 25:33-9. [PMID: 26476955 DOI: 10.1016/j.carpath.2015.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/02/2015] [Accepted: 09/20/2015] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Complement activation contributes significantly to inflammation-related damage in the heart after acute myocardial infarction. Knowledge on factors that regulate postinfraction complement activation is incomplete however. In this study, we investigated whether endogenous C1-inhibitor, a well-known inhibitor of complement activation, is expressed in the heart after acute myocardial infarction. MATERIALS AND METHODS C1-inhibitor and complement activation products C3d and C4d were analyzed immunohistochemically in the hearts of patients who died at different time intervals after acute myocardial infarction (n=28) and of control patients (n=8). To determine putative local C1-inhibitor production, cardiac transcript levels of the C1-inhibitor-encoding gene serping1 were determined in rats after induction of acute myocardial infarction (microarray). Additionally, C1-inhibitor expression was analyzed (fluorescence microscopy) in human endothelial cells and rat cardiomyoblasts in vitro. RESULTS C1-inhibitor was found predominantly in and on jeopardized cardiomyocytes in necrotic infarct cores between 12h and 5days old. C1-inhibitor protein expression coincided in time and colocalized with C3d and C4d. In the rat heart, serping1 transcript levels were increased from 2h up until 7days after acute myocardial infarction. Both endothelial cells and cardiomyoblasts showed increased intracellular expression of C1-inhibitor in response to ischemia in vitro (n=4). CONCLUSIONS These observations suggest that endogenous C1-inhibitor is likely involved in the regulation of complement activity in the myocardium following acute myocardial infarction. Observations in rat and in vitro suggest that C1-inhibitor is produced locally in the heart after acute myocardial infarction.
Collapse
|
21
|
Ma Y, Kong L, Nan K, Qi S, Ru L, Ding C, Wang D. Apolipoprotein-J prevents angiotensin II-induced apoptosis in neonatal rat ventricular cells. Lipids Health Dis 2015; 14:114. [PMID: 26391229 PMCID: PMC4578334 DOI: 10.1186/s12944-015-0118-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/10/2015] [Indexed: 11/22/2022] Open
Abstract
Background Up-regulation of angiotensin II (AngII) occurs in cardiac diseases, such as congestive heart failure, cardiac hypertrophy, myocardial ischemia and atrial fibrillation, which represent major health problems. Evidence from in vivo studies suggests that the level of Apolipoprotein-J (ApoJ) is also elevated but plays a protective role in cardiovascular disease. This study aimed to evaluate the protective effects of ApoJ against cytotoxicity of AngII in neonatal rat ventricular cells (NRVCs). Methods and results In culture, NRVCs were damaged by exposure to AngII, and ApoJ overexpression using an adenovirus vector significantly reduced the AngII-induced cell injury. ApoJ also prevented AngII from augmenting Nox2/gp91phox expression. The reactive oxygen species (ROS) scavenger, Mn(III)TBAP, showed similar results of attenuating AngII-induced cell damage. Furthermore, ApoJ overexpression increased phosphorylation of Akt, and the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 diminished the antioxidant effects of ApoJ, and prevented the protective effect of ApoJ against the cytotoxicity of AngII. Moreover, upregulation of nuclear factor κB (NF-κB) p65 expression and phosphorylation of p38 mitogen-activated protein kinase (MAPK) mediated by AngII in cultured NRVCs were significantly inhibited by overexpression of ApoJ. The p38 MAPK inhibitor SB203580 and the NF-κB inhibitor PDTC protected NRVCs from injury caused by AngII. Conclusions ApoJ serves as a cytoprotective protein in NRVCs against cytotoxicity of AngII through the PI3K-Akt-ROS and MAPK/ NF-κB pathways.
Collapse
Affiliation(s)
- Yanzhuo Ma
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Lingfeng Kong
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Kai Nan
- Health and Medical Development Research Center of Hebei Province, Shijiazhuang, Hebei, China.
| | - Shuying Qi
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Leisheng Ru
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Chao Ding
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Dongmei Wang
- Department of Cardiology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
22
|
Lymphocytes Infiltrate the Quadriceps Muscle in Lymphocytic Myocarditis Patients: A Potential New Diagnostic Tool. Can J Cardiol 2014; 30:1547-54. [DOI: 10.1016/j.cjca.2014.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 11/21/2022] Open
|
23
|
Proteomic analysis allows for early detection of potential markers of metabolic impairment in very young obese children. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2014; 2014:9. [PMID: 24949022 PMCID: PMC4063220 DOI: 10.1186/1687-9856-2014-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/04/2014] [Indexed: 12/22/2022]
Abstract
Background Early diagnosis of initial metabolic derangements in young obese children could influence their management; however, this impairment is frequently not overt, but subtle and undetectable by routinely used clinical assays. Our aim was to evaluate the ability of serum proteomic analysis to detect these incipient metabolic alterations in comparison to standard clinical methods and to identify new candidate biomarkers. Methods A cross-sectional study of fasting serum samples from twenty-two prepubertal, Caucasian obese (OB; 9.22 ± 1.93 years; 3.43 ± 1.08 BMI-SDS) and twenty-one lean controls (C; 8.50 ± 1.98 years; -0.48 ± 0.81 BMI-SDS) and a prospective study of fasting serum samples from twenty prepubertal, Caucasian obese children (11 insulin resistant [IR]) before (4.77 ± 1.30 BMI-SDS) and after weight reduction (2.57 ± 1.29 BMI-SDS) by conservative treatment in a reference hospital (Pros-OB) was performed. Proteomic analysis (two-dimension-eletrophoresis + mass spectrometry analysis) of serum and comparative evaluation of the sensitivity of routinely used assays in the clinics to detect the observed differences in protein expression level, as well as their relationship with anthropometric features, insulin resistance indexes, lipid profile and adipokine levels were carried out. Results Study of the intensity data from proteomic analysis showed a decrease of several isoforms of apolipoprotein-A1, apo-J/clusterin, vitamin D binding protein, transthyretin in OBvs. C, with some changes in these proteins being enhanced by IR and partially reversed after weight loss. Expression of low molecular weight isoforms of haptoglobin was increased in OB, enhanced in IR and again decreased after weight loss, being positively correlated with serum interleukin-6 and NAMPT/visfatin levels. After statistical correction for multiple comparisons, significance remained for a single isoform of low MW haptoglobin (OB vs. C and IR vs. non-IR) and Apo A1 (IR vs. non-IR). Assays routinely used in the clinical setting (ELISA/kinetic nephelometry), only partially confirmed the changes observed by proteomic analysis (ApoA1 and haptoglobin). Conclusion Proteomic analysis can allow for the identification of potential new candidate biomarkers as a complement to routinely used assays to detect initial changes in serum markers of inflammation and lipid metabolism impairment in young obese children.
Collapse
|
24
|
Carrano A, Snkhchyan H, Kooij G, van der Pol S, van Horssen J, Veerhuis R, Hoozemans J, Rozemuller A, de Vries HE. ATP-binding cassette transporters P-glycoprotein and breast cancer related protein are reduced in capillary cerebral amyloid angiopathy. Neurobiol Aging 2014; 35:565-75. [DOI: 10.1016/j.neurobiolaging.2013.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 08/30/2013] [Accepted: 09/10/2013] [Indexed: 12/12/2022]
|
25
|
Mulder SD, Nielsen HM, Blankenstein MA, Eikelenboom P, Veerhuis R. Apolipoproteins E and J interfere with amyloid-beta uptake by primary human astrocytes and microglia in vitro. Glia 2014; 62:493-503. [PMID: 24446231 DOI: 10.1002/glia.22619] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 11/30/2013] [Accepted: 12/06/2013] [Indexed: 12/12/2022]
Abstract
Defective clearance of the amyloid-β peptide (Aβ) from the brain is considered a strong promoter in Alzheimer's disease (AD) pathogenesis. Astrocytes and microglia are important mediators of Aβ clearance and Aβ aggregation state and the presence of amyloid associated proteins (AAPs), such as Apolipoproteins E and J (ApoE and ApoJ), may influence Aβ clearance by these cells. Here we set out to investigate whether astrocytes and microglia differ in uptake efficiency of Aβ oligomers (Aβoligo ) and Aβ fibrils (Aβfib ), and whether the Aβ aggregation state and/or presence of AAPs affect Aβ uptake in these cells in vitro. Adult human primary microglia and astrocytes, isolated from short delay post-mortem brain tissue, were exposed to either Aβoligo or Aβfib alone or combined with a panel of certain AAPs whereafter Aβ-positive cells were quantified using flow cytometry. Upon exposure to Aβ combined with ApoE, ApoJ, α1-antichymotrypsin (ACT) and a combination of serum amyloid P and complement C1q (SAP-C1q), a clear reduction in astrocytic but not microglial Aβoligo uptake, was observed. In contrast, Aβfib uptake was strongly reduced in the presence of AAPs in microglia, but not in astrocytes. These data provide the first evidence of distinct roles of microglia and astrocytes in Aβ clearance. More importantly we show that Aβ clearance by glial cells is negatively affected by AAPs like ApoE and ApoJ. Thus, targeting the association of Aβ with AAPs, such as ApoE and ApoJ, could serve as a therapeutic strategy to increase Aβ clearance by glial cells.
Collapse
Affiliation(s)
- Sandra D Mulder
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands; Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
26
|
Plasma clusterin concentrations may predict resistance to intravenous immunoglobulin in patients with Kawasaki disease. ScientificWorldJournal 2013; 2013:382523. [PMID: 23956692 PMCID: PMC3727184 DOI: 10.1155/2013/382523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/05/2013] [Indexed: 12/04/2022] Open
Abstract
Kawasaki disease (KD) is an acute febrile vasculitic syndrome of early childhood often complicated by coronary artery lesion that drastically reduces the quality of life. The study aimed to identify a reliable marker for predicting nonresponsiveness to the first course of intravenous immunoglobulin (IVIG) in KD patients. A total of 63 patients with KD were enrolled in the study (IVIG response, 58; IVIG resistance, 5). Plasma samples were collected before and after IVIG infusion for measurement of biomarkers. Patients' clinical characteristics and laboratory data were also analyzed. A receiver operating characteristic curve was generated to identify a cut-off value for predicting IVIG resistance. Among the biomarkers, the difference in plasma clusterin concentrations before and after IVIG infusion (CLUSTER 12) was significantly related to IVIG resistance (P = 0.040; 95% confidence interval (CI): −25.8% to −6.0%). Using a CLUSTER 12 cut-off value of <8.52 mg/L, the odds ratio for IVIG resistance was 11.467 (95% CI: 1.186 to 110.853). Patients with plasma CLUSTER 12 concentrations >8.52 mg/L had a much higher risk of IVIG resistance than those with CLUSTER 12 concentrations <8.52 mg/L. Plasma clusterin concentration shows promise as a candidate biomarker for predicting IVIG resistance in patients with KD.
Collapse
|
27
|
van Dijk A, Naaijkens BA, Jurgens WJFM, Oerlemans R, Scheffer GL, Kassies J, Aznou J, Brouwer M, van Rossum AC, Schuurhuis GJ, van Milligen FJ, Niessen HWM. The multidrug resistance protein breast cancer resistance protein (BCRP) protects adipose-derived stem cells against ischemic damage. Cell Biol Toxicol 2012; 28:303-15. [PMID: 22801743 DOI: 10.1007/s10565-012-9225-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 07/02/2012] [Indexed: 12/29/2022]
Abstract
Adipose tissue-derived stem cells (ASCs) are promising candidates for regenerative therapy, like after myocardial infarction. However, when transplanted into the infarcted heart, ASCs are jeopardized by the ischemic environment. Interestingly, it has been shown that multidrug resistance (MDR) proteins like the breast cancer resistance protein (BCRP) and P-glycoprotein (P-gp) have a protective effect in haematopoietic stem cells. In ASC, however, only expression of BCRP was shown until now. In this study, we therefore analysed the expression and functional activity of BCRP and P-gp and their putative function in ischemia in ASC. BCRP and P-gp protein expression was studied over time (passages 2-6) using western blot analysis and immunohistochemical staining. MDR activity was analysed using protein-specific substrate extrusion assays. Ischemia was induced using metabolic inhibition. All analyses demonstrated protein expression and activity of BCRP in ASCs. In contrast, only minor expression of P-gp was found, without functional activity. BCRP expression was most prominent in early passage ASCs (p2) and decreased during culture. Finally, ischemia induced expression of BCRP. In addition, when BCRP was blocked, a significant increase in dead ASCs was found already after 1 h of ischemia. In conclusion, ASCs expressed BCRP, especially in early passages. In addition, we now show for the first time that BCRP protects ASCs against ischemia-induced cell death. These data therefore indicate that for transplantation of ASCs in an ischemic environment, like myocardial infarction, the optimal stem cell protective effect of BCRP theoretically will be achieved with early culture passages ASCs.
Collapse
Affiliation(s)
- A van Dijk
- Department of Pathology, VU University Medical Centre, Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yanaba K, Asano Y, Tada Y, Sugaya M, Kadono T, Sato S. A possible contribution of elevated serum clusterin levels to the inhibition of digital ulcers and pulmonary arterial hypertension in systemic sclerosis. Arch Dermatol Res 2012; 304:459-63. [DOI: 10.1007/s00403-012-1219-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/22/2012] [Accepted: 01/26/2012] [Indexed: 11/30/2022]
|
29
|
Gupta S, Halushka MK, Hilton GM, Arking DE. Postmortem cardiac tissue maintains gene expression profile even after late harvesting. BMC Genomics 2012; 13:26. [PMID: 22251372 PMCID: PMC3342086 DOI: 10.1186/1471-2164-13-26] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/17/2012] [Indexed: 11/24/2022] Open
Abstract
Background Gene expression studies can be used to help identify disease-associated genes by comparing the levels of expressed transcripts between cases and controls, and to identify functional genetic variants (expression quantitative loci or eQTLs) by comparing expression levels between individuals with different genotypes. While many of these studies are performed in blood or lymphoblastoid cell lines due to tissue accessibility, the relevance of expression differences in tissues that are not the primary site of disease is unclear. Further, many eQTLs are tissue specific. Thus, there is a clear and compelling need to conduct gene expression studies in tissues that are specifically relevant to the disease of interest. One major technical concern about using autopsy-derived tissue is how representative it is of physiologic conditions, given the effect of postmortem interval on tissue degradation. Results In this study, we monitored the gene expression of 13 tissue samples harvested from a rapid autopsy heart (non-failed heart) and 7 from a cardiac explant (failed heart) through 24 hours of autolysis. The 24 hour autopsy simulation was designed to reflect a typical autopsy scenario where a body may begin cooling to ambient temperature for ~12 hours, before transportation and storage in a refrigerated room in a morgue. In addition, we also simulated a scenario wherein the body was left at room temperature for up to 24 hours before being found. A small fraction (< 2.5%) of genes showed fluctuations in expression over the 24 hr period and largely belong to immune and signal response and energy metabolism-related processes. Global expression analysis suggests that RNA expression is reproducible over 24 hours of autolysis with 95% genes showing < 1.2 fold change. Comparing the rapid autopsy to the failed heart identified 480 differentially expressed genes, including several types of collagens, lumican (LUM), natriuretic peptide A (NPPA) and connective tissue growth factor (CTGF), which allows for the clear separation between failing and non-failing heart based on gene expression profiles. Conclusions Our results demonstrate that RNA from autopsy-derived tissue, even up to 24 hours of autolysis, can be used to identify biologically relevant expression pattern differences, thus serving as a practical source for gene expression experiments.
Collapse
Affiliation(s)
- Simone Gupta
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
30
|
Mulder SD, Veerhuis R, Blankenstein MA, Nielsen HM. The effect of amyloid associated proteins on the expression of genes involved in amyloid-β clearance by adult human astrocytes. Exp Neurol 2012; 233:373-9. [DOI: 10.1016/j.expneurol.2011.11.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 10/25/2011] [Accepted: 11/01/2011] [Indexed: 11/24/2022]
|
31
|
Takase O, Hishikawa K, Kamiura N, Nakakuki M, Kawano H, Mizuguchi K, Fujita T. Eicosapentaenoic acid regulates IκBα and prevents tubulointerstitial injury in kidney. Eur J Pharmacol 2011; 669:128-35. [PMID: 21871881 DOI: 10.1016/j.ejphar.2011.07.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 07/18/2011] [Accepted: 07/29/2011] [Indexed: 11/28/2022]
Abstract
Fish oil containing n-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is well known to prevent the progression of IgA nephropathy. However, the mechanism through which fish oil prevents the progression of renal injury remains uncertain. We tried to clarify the effects of EPA on tubulointerstitial injury in the kidney both in vivo and in vitro. We examined the effects of EPA, especially to focus on nuclear factor kappa B (NF-κB), using Thy-1 nephritis models. Also the mechanism of EPA was investigated using small-interfering RNA (siRNA) in lipopolysaccharide (LPS)-stimulated proximal tubular epithelial cells (PTECs). In Thy-1 nephritis models, EPA significantly inhibited tubulointerstitial injury and the infiltration of macrophages into tubulointerstitial lesions except severe glomerular injury at early stage. Compared with control animals, NF-κB activation was significantly augmented in the Thy-1 nephritic kidney. However, treatment with EPA significantly reduced NF-κB activation, down-regulated the expressions of NF-κB-dependent molecules. Also in LPS-stimulated PTECs, LPS augmented NF-κB activation and the expression of NF-κB-dependent molecules. As in the case with the Thy-1 nephritis models, treatment with EPA inhibited them, prevented the degradation of IκBα in LPS-stimulated PTECs. Pre-treatment with siRNA for IκBα abolished the inhibitory effect of EPA on LPS-induced NF-κB activation, suggesting that EPA inhibited NF-κB activation by regulating IκBα. Our results indicate that EPA prevents the early progression of tubulointerstitial injury in Thy-1 nephritis models, and the inhibitory effect of EPA on the expression of inflammatory molecules via the regulation of IκBα in cultured cells may explain this mechanism.
Collapse
Affiliation(s)
- Osamu Takase
- Department of Clinical Renal Regeneration, and Division of Nephrology and Endocrinology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Sipkens JA, Hahn NE, van Nieuw-Amerongen GP, Stehouwer CDA, Rauwerda JA, van Hinsbergh VWM, Niessen HWM, Krijnen PAJ. Homocysteine induces phosphatidylserine exposure in cardiomyocytes through inhibition of Rho kinase and flippase activity. Cell Physiol Biochem 2011; 28:53-62. [PMID: 21865848 DOI: 10.1159/000331713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2011] [Indexed: 01/05/2023] Open
Abstract
AIMS Increased levels of homocysteine (Hcy) form an independent risk factor for cardiovascular disease. In a previous study we have shown that Hcy induced phosphatidylserine (PS) exposure to the outer leaflet of the plasma membrane in cardiomyocytes, inducing a pro-inflammatory phenotype. In the present study the mechanism(s) involved in Hcy-induced PS exposure were analyzed. METHODS H9c2 rat cardiomyoblasts were subjected to 2.5 mM D,L-Hcy and analyzed for RhoA translocation and activity, Rho Kinase (ROCK) activity and expression and flippase activity. In addition, the effect of ROCK inhibition with Y27632 on Hcy-induced PS exposure and flippase activity was analyzed. Furthermore, GTP and ATP levels were determined. RESULTS Incubation of H9c2 cells with 2.5 mM D,L-Hcy did not inhibit RhoA translocation to the plasma membrane. Neither did it inhibit activation of RhoA, even though GTP levels were significantly decreased. Hcy did significantly inhibit ROCK activation, but not its expression, and did inhibit flippase activity, in advance of a significant decrease in ATP levels. ROCK inhibition via Y27632 did not have significant added effects on this. CONCLUSION Hcy induced PS exposure in the outer leaflet of the plasma membrane in cardiomyocytes via inhibition of ROCK and flippase activity. As such Hcy may induce cardiomyocytes vulnerable to inflammation in vivo in hyperhomocysteinaemia patients.
Collapse
Affiliation(s)
- Jessica A Sipkens
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hahn NE, Meischl C, Wijnker PJM, Musters RJP, Fornerod M, Janssen HWRM, Paulus WJ, van Rossum AC, Niessen HWM, Krijnen PAJ. NOX2, p22phox and p47phox are targeted to the nuclear pore complex in ischemic cardiomyocytes colocalizing with local reactive oxygen species. Cell Physiol Biochem 2011; 27:471-8. [PMID: 21691064 DOI: 10.1159/000329968] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2011] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND NADPH oxidases play an essential role in reactive oxygen species (ROS)-based signaling in the heart. Previously, we have demonstrated that (peri)nuclear expression of the catalytic NADPH oxidase subunit NOX2 in stressed cardiomyocytes, e.g. under ischemia or high concentrations of homocysteine, is an important step in the induction of apoptosis in these cells. Here this ischemia-induced nuclear targeting and activation of NOX2 was specified in cardiomyocytes. METHODS The effect of ischemia, mimicked by metabolic inhibition, on nuclear localization of NOX2 and the NADPH oxidase subunits p22(phox) and p47(phox), was analyzed in rat neonatal cardiomyoblasts (H9c2 cells) using Western blot, immuno-electron microscopy and digital-imaging microscopy. RESULTS NOX2 expression significantly increased in nuclear fractions of ischemic H9c2 cells. In addition, in these cells NOX2 was found to colocalize in the nuclear envelope with nuclear pore complexes, p22(phox), p47(phox) and nitrotyrosine residues, a marker for the generation of ROS. Inhibition of NADPH oxidase activity, with apocynin and DPI, significantly reduced (peri)nuclear expression of nitrotyrosine. CONCLUSION We for the first time show that NOX2, p22(phox) and p47(phox) are targeted to and produce ROS at the nuclear pore complex in ischemic cardiomyocytes.
Collapse
Affiliation(s)
- Nynke E Hahn
- Department of Pathology, VU University Medical Centre, De Boelelaan 1117, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Jun HO, Kim DH, Lee SW, Lee HS, Seo JH, Kim JH, Kim JH, Yu YS, Min BH, Kim KW. Clusterin protects H9c2 cardiomyocytes from oxidative stress-induced apoptosis via Akt/GSK-3β signaling pathway. Exp Mol Med 2011; 43:53-61. [PMID: 21270507 DOI: 10.3858/emm.2011.43.1.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Clusterin is a secretory glycoprotein, which is highly up-regulated in a variety of normal and injury tissues undergoing apoptosis including infarct region of the myocardium. Here, we report that clusterin protects H9c2 cardiomyocytes from H2O2-induced apoptosis by triggering the activation of Akt and GSK-3β. Treatment with H2O2 induces apoptosis of H9c2 cells by promoting caspase cleavage and cytochrome c release from mitochondria. However, co-treatment with clusterin reverses the induction of apoptotic signaling by H2O2, thereby recovers cell viability. The protective effect of clusterin on H2O2-induced apoptosis is impaired by PI3K inhibitor LY294002, which effectively suppresses clusterin-induced activation of Akt and GSK-3β. In addition, the protective effect of clusterin is independent on its receptor megalin, because inhibition of megalin has no effect on clusterin-mediated Akt/GSK-3β phosphoylation and H9c2 cell viability. Collectively, these results suggest that clusterin has a role protecting cardiomyocytes from oxidative stress and the Akt/GSK-3β signaling mediates anti-apoptotic effect of clusterin.
Collapse
Affiliation(s)
- Hyoung-Oh Jun
- NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Singla DK, Singla RD, Lamm S, Glass C. TGF-β2 treatment enhances cytoprotective factors released from embryonic stem cells and inhibits apoptosis in infarcted myocardium. Am J Physiol Heart Circ Physiol 2011; 300:H1442-50. [PMID: 21297031 DOI: 10.1152/ajpheart.00917.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated whether factors released from mouse embryonic stem (ES) cells primed with and without transforming growth factor (TGF)-β2 inhibit iodoacetic acid (IAA)- and H(2)O(2)-induced apoptosis in the cell culture system as well as after transplantation in the infarcted heart. We generated conditioned media (CMs) from ES cells primed with and without TGF-β2 and determined their effects on IAA- and H(2)O(2)-induced apoptosis in H9c2 cells. We also transplanted both ES-CMs in the infarcted heart to determine the effects on apoptosis and cardiac function after myocardial infarction (MI) at day (D)1 and D14. Terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL) staining, apoptotic ELISA, and cell viability data demonstrated significantly (P < 0.05) reduced apoptosis with ES-CM compared with controls in both cell culture models. Moreover, TGF-β2-primed ES-CM (T-ES-CM) demonstrated enhanced beneficial effects, with further reduced (P < 0.05) apoptosis compared with ES-CM, suggesting the a presence of additional cytoprotective released factors after TGF-β2 treatment. Next, our in vivo apoptosis data suggested significant decrease in apoptosis with both ES-CMs compared with MI alone at D1 and D14. Notably, T-ES-CM demonstrated significant (P < 0.05) inhibition of apoptosis and fibrosis with improved cardiac function compared with ES-CM at D14, whereas no such effects were observed at D1. Next, we confirmed that apoptosis is mediated through a prosurvival Akt pathway. Moreover, we determined that after TGF-β2 treatment there was a two- to fivefold increase in cytoprotective released factors (interleukin-10, stem cell factor, tissue inhibitor of matrix metalloproteinase-1, and VEGF) with T-ES-CM compared with ES-CM. In conclusion, we suggest that factors released from ES cells with and without TGF-β2 treatment contain antiapoptotic factors that inhibit apoptosis in vitro and in vivo. We also suggest that T-ES-CM demonstrates additional beneficial effects that provide useful information for future therapeutic applications in regenerative medicine.
Collapse
Affiliation(s)
- Dinender K Singla
- Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, 32816, USA.
| | | | | | | |
Collapse
|
36
|
Greene MJ, Sam F, Soo Hoo PT, Patel RS, Seldin DC, Connors LH. Evidence for a functional role of the molecular chaperone clusterin in amyloidotic cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:61-8. [PMID: 21224044 DOI: 10.1016/j.ajpath.2010.11.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/02/2010] [Accepted: 09/30/2010] [Indexed: 01/20/2023]
Abstract
Molecular chaperones, including the extracellular protein clusterin (CLU), play a significant role in maintaining proteostasis; they have a unique capacity to bind and stabilize non-native protein conformations, prevent aggregation, and keep proteins in a soluble folding-competent state. In this study, we investigated amyloid-infiltrated cardiac tissue for the presence of CLU and measured serum levels of CLU in patients with and without amyloidotic cardiomyopathy (CMP). Cardiac tissues containing amyloid deposits composed of either transthyretin (TTR) or Ig light chain from nine patients with amyloidotic CMP were examined for the presence of CLU using immunohistochemical techniques. CLU staining coincided with the extracellular myocardial amyloid deposits in tissues from patients with familial TTR, senile systemic, and Ig light chain amyloidosis. The association of CLU with cardiac amyloid deposits was confirmed by immunogold electron microscopy. Serum concentrations of CLU were measured in familial TTR, senile systemic, and Ig light chain amyloidosis patient groups and compared with both age-matched healthy controls and with patients with CMP unrelated to amyloid disease. Subset analysis of disease cohorts, based on cardiac involvement, indicated that decreased serum CLU concentrations were associated with amyloidotic CMP. Taken together, these results suggest that CLU may play a pathogenetic role in TTR and Ig light chain amyloidoses and amyloidotic CMP.
Collapse
Affiliation(s)
- Michael J Greene
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
37
|
Van Dijk A, Vermond RA, Krijnen PAJ, Juffermans LJM, Hahn NE, Makker SP, Aarden LA, Hack E, Spreeuwenberg M, van Rossum BC, Meischl C, Paulus WJ, Van Milligen FJ, Niessen HWM. Intravenous clusterin administration reduces myocardial infarct size in rats. Eur J Clin Invest 2010; 40:893-902. [PMID: 20854280 DOI: 10.1111/j.1365-2362.2010.02345.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Clusterin (Apolipoprotein J), a plasma protein with cytoprotective and complement-inhibiting activities, localizes in the infarcted heart during myocardial infarction (MI). Recently, we have shown a protective effect of exogenous clusterin in vitro on ischaemically challenged cardiomyocytes independent of complement. We therefore hypothesized that intravenous clusterin administration would reduce myocardial infarction damage. METHODS Wistar rats undergoing experimental MI, induced by 40 min ligation of a coronary vessel, were treated with either clusterin (n=15) or vehicle (n=13) intravenously, for 3 days post-MI. After 4 weeks, hearts were analysed. The putative role of megalin, a clusterin receptor, was also studied. RESULTS Administration of human clusterin significantly reduced both infarct size (with 75 ± 5%) and death of animals (23% vehicle group vs. 0% clusterin group). Importantly, histochemical analysis showed no signs of impaired wound healing in the clusterin group. In addition, significantly increased numbers of macrophages were found in the clusterin group. We also found that the clusterin receptor megalin was present on cardiomyocytes in vitro which, however, was not influenced by ischaemia. Human clusterin co-localized with this receptor in vitro, but not in the human heart. In addition, using a megalin inhibitor, we found that clusterin did not exert its protective effect on cardiomyocytes through megalin. CONCLUSIONS Our results thus show that clusterin has a protective effect on cardiomyocytes after acute myocardial infarction in vivo, independent of its receptor megalin. This indicates that clusterin, or a clusterin derivate, is a potential therapeutic agent in the treatment of MI.
Collapse
|
38
|
Nielsen HM, Mulder SD, Beliën JAM, Musters RJP, Eikelenboom P, Veerhuis R. Astrocytic A beta 1-42 uptake is determined by A beta-aggregation state and the presence of amyloid-associated proteins. Glia 2010; 58:1235-46. [PMID: 20544859 DOI: 10.1002/glia.21004] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Intracerebral accumulation of amyloid-beta (A beta) leading to A beta plaque formation, is the main hallmark of Alzheimer's disease and might be caused by defective A beta-clearance. We previously found primary human astrocytes and microglia able to bind and ingest A beta 1-42 in vitro, which appeared to be limited by A beta 1-42 fibril formation. We now confirm that astrocytic A beta-uptake depends on size and/or composition of A beta-aggregates as astrocytes preferably take up oligomeric A beta over fibrillar A beta. Upon exposure to either fluorescence-labelled A beta 1-42 oligomers (A beta(oligo)) or fibrils (A beta(fib)), a larger (3.7 times more) proportion of astrocytes ingested oligomers compared to fibrils, as determined by flow cytometry. A beta-internalization was verified using confocal microscopy and live-cell imaging. Neither uptake of A beta(oligo) nor A beta(fib), triggered proinflammatory activation of the astrocytes, as judged by quantification of interleukin-6 and monocyte-chemoattractant protein-1 release. Amyloid-associated proteins, including alpha1-antichymotrypsin (ACT), serum amyloid P component (SAP), C1q and apolipoproteins E (ApoE) and J (ApoJ) were earlier found to influence A beta-aggregation. Here, astrocytic uptake of A beta(fib) increased when added to the cells in combination with SAP and C1q (SAP/C1q), but was unchanged in the presence of ApoE, ApoJ and ACT. Interestingly, ApoJ and ApoE dramatically reduced the number of A beta(oligo)-positive astrocytes, whereas SAP/C1q slightly reduced A beta(oligo) uptake. Thus, amyloid-associated proteins, especially ApoJ and ApoE, can alter A beta-uptake in vitro and hence may influence A beta clearance and plaque formation in vivo.
Collapse
Affiliation(s)
- Henrietta M Nielsen
- Department of Clinical Chemistry, VU University Medical Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
39
|
Li Y, Qu J, Shelat H, Gao S, Wassler M, Geng YJ. Clusterin induces CXCR4 expression and migration of cardiac progenitor cells. Exp Cell Res 2010; 316:3435-42. [PMID: 20813109 DOI: 10.1016/j.yexcr.2010.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 08/20/2010] [Accepted: 08/23/2010] [Indexed: 11/18/2022]
Abstract
Clusterin (CST) is a stress-responding protein with multiple biological functions, including the inhibition of apoptosis and inflammation and transport of lipids. It may also participate in cell traffic and migration. In the process of post-infarct cardiac tissue repair, stem cells migrate into the damaged myocardium under the influence of chemoattractive substances such as stromal cell-derived factor (SDF). This study aimed at testing whether CST enhances expression of stem cell homing receptor and migration of cardiac progenitor cells (CPCs). CPCs isolated from fetal canine hearts transduced by CST cDNA expressed high levels of CXCR4, a receptor for SDF-1. The transfected cells also showed an increased migratory response to SDF-1 stimulation. The SDF-1-mediated migration of the CST-expressing CPCs was attenuated by PI3 kinase inhibitor LY294002 but not by mitogen-activated protein/ERK kinase inhibitor PD98059. Analysis of cell cycle by flow cytometry revealed no significant difference in cell cycle between the transduced and control CPCs. Thus, CST expression may increase CPCs migration via increasing CXCR4 expression and SDF-1/chemokine receptor signaling in a PI3/Akt-dependent manner.
Collapse
Affiliation(s)
- Yangxin Li
- Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Inhibition of Rho-ROCK signaling induces apoptotic and non-apoptotic PS exposure in cardiomyocytes via inhibition of flippase. J Mol Cell Cardiol 2010; 49:781-90. [PMID: 20691698 DOI: 10.1016/j.yjmcc.2010.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 07/12/2010] [Accepted: 07/28/2010] [Indexed: 02/06/2023]
Abstract
Subsequent to myocardial infarction, cardiomyocytes within the infarcted areas and border zones expose phosphatidylserine (PS) in the outer plasma membrane leaflet (flip-flop). We showed earlier that in addition to apoptosis, this flip-flop can be reversible in cardiomyocytes. We now investigated a possible role for Rho and downstream effector Rho-associated kinase (ROCK) in the process of (reversible) PS exposure and apoptosis in cardiomyocytes. In rat cardiomyoblasts (H9c2 cells) and isolated adult ventricular rat cardiomyocytes Clostridium difficile Toxin B (TcdB), a Rho GTPase family inhibitor, C3 transferase (C3), a Rho(A,B,C) inhibitor and the ROCK inhibitors Y27632 and H1152 were used to inhibit Rho-ROCK signaling. PS exposure was assessed via flow cytometry and fluorescent digital imaging microscopy using annexin V. Akt expression and phosphorylation were analyzed via Western blot, and Akt activity was inhibited by wortmannin. The cellular concentration activated caspase 3 was determined as a measure of apoptosis, and flippase activity was assessed via flow cytometry using NBD-labeled PS. TcdB, C3, Y27632 and H1152 all significantly increased PS exposure. TcdB, Y27632 and H1152 all significantly inhibited phosphorylation of the anti-apoptotic protein Akt and Akt inhibition by wortmannin lead to increased PS exposure. However, only TcdB and C3, but not ROCK- or Akt inhibition led to caspase 3 activation and thus apoptosis. Notably, pancaspase inhibitor zVAD only partially inhibited TcdB-induced PS exposure indicating the existence of apoptotic and non-apoptotic PS exposure. The induced PS exposure coincided with decreased flippase activity as measured with NBD-labeled PS flip-flop. In this study, we show a regulatory role for a novel signaling route, Rho-ROCK-flippase signaling, in maintaining asymmetrical membrane phospholipid distribution in cardiomyocytes.
Collapse
|
41
|
Chapter 9: Oxidative stress in malignant progression: The role of Clusterin, a sensitive cellular biosensor of free radicals. Adv Cancer Res 2010; 104:171-210. [PMID: 19878777 DOI: 10.1016/s0065-230x(09)04009-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clusterin/Apolipoprotein J (CLU) gene is expressed in most human tissues and encodes for two protein isoforms; a conventional heterodimeric secreted glycoprotein and a truncated nuclear form. CLU has been functionally implicated in several physiological processes as well as in many pathological conditions including ageing, diabetes, atherosclerosis, degenerative diseases, and tumorigenesis. A major link of all these, otherwise unrelated, diseases is that they are characterized by increased oxidative injury due to impaired balance between production and disposal of reactive oxygen or nitrogen species. Besides the aforementioned diseases, CLU gene is differentially regulated by a wide variety of stimuli which may also promote the production of reactive species including cytokines, interleukins, growth factors, heat shock, radiation, oxidants, and chemotherapeutic drugs. Although at low concentration reactive species may contribute to normal cell signaling and homeostasis, at increased amounts they promote genomic instability, chronic inflammation, lipid oxidation, and amorphous aggregation of target proteins predisposing thus cells for carcinogenesis or other age-related disorders. CLU seems to intervene to these processes due to its small heat-shock protein-like chaperone activity being demonstrated by its property to inhibit protein aggregation and precipitation, a main feature of oxidant injury. The combined presence of many potential regulatory elements in the CLU gene promoter, including a Heat-Shock Transcription Factor-1 and an Activator Protein-1 element, indicates that CLU gene is an extremely sensitive cellular biosensor of even minute alterations in the cellular oxidative load. This review focuses on CLU regulation by oxidative injury that is the common molecular link of most, if not all, pathological conditions where CLU has been functionally implicated.
Collapse
|
42
|
ter Weeme M, Vonk ABA, Kupreishvili K, van Ham M, Zeerleder S, Wouters D, Stooker W, Eijsman L, Van Hinsbergh VWM, Krijnen PAJ, Niessen HWM. Activated complement is more extensively present in diseased aortic valves than naturally occurring complement inhibitors: a sign of ongoing inflammation. Eur J Clin Invest 2010; 40:4-10. [PMID: 19843156 DOI: 10.1111/j.1365-2362.2009.02216.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recent studies indicate a role for complement in the pathogenesis of aortic valve disease. However, the role of naturally occurring anti-complement mediators in this context is unknown. In this study, we have analysed this in three different pathological conditions of the aortic valve: degeneration, atherosclerosis and bacterial endocarditis. MATERIALS AND METHODS Human aortic valves were obtained at autopsy (n = 30): 5 control valves, 10 aortic valves with atherosclerotic changes, 10 aortic valves with degenerative changes and 5 degenerative changed aortic valves with bacterial infection. These valves were analysed immunohistochemically for the presence of activated complement (C3d and C5b9) and the complement inhibitors C1-inh and clusterin. Areas of positivity were then quantified. RESULTS C3d, C5b9 and the complement inhibitors C1-inh and clusterin depositions were mainly found in the endothelium and extracellular matrix in aortic valves. All these mediators were already present in control valves, but the area of positivity increased significantly in response to the different diseases, with the highest increase in response to bacterial endocarditis. Interestingly, in all three aortic diseases, the depositions of complement were significantly more widespread than that of their inhibitors. CONCLUSIONS Our study indicates that anti-complement mediators (C1-inh and clusterin) are deposited in diseased aortic valves together with activated complement, indicating an existing counter response against complement locally in the valve. However, deposition of activated complement is significantly more widespread than that of its inhibitors, which could explain ongoing inflammation in those diseased aortic valves.
Collapse
|
43
|
van Dijk A, Krijnen PAJ, Vermond RA, Pronk A, Spreeuwenberg M, Visser FC, Berney R, Paulus WJ, Hack CE, van Milligen FJ, Niessen HWM. Inhibition of type 2A secretory phospholipase A2 reduces death of cardiomyocytes in acute myocardial infarction. Apoptosis 2009; 14:753-63. [DOI: 10.1007/s10495-009-0350-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Urashima T, Zhao M, Wagner R, Fajardo G, Farahani S, Quertermous T, Bernstein D. Molecular and physiological characterization of RV remodeling in a murine model of pulmonary stenosis. Am J Physiol Heart Circ Physiol 2008; 295:H1351-H1368. [PMID: 18586894 DOI: 10.1152/ajpheart.91526.2007] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Right ventricular (RV) dysfunction is a common long-term complication in patients after the repair of congenital heart disease. Previous investigators have examined the cellular and molecular mechanisms of left ventricular (LV) remodeling, but little is known about the stressed RV. Our purpose was to provide a detailed physiological characterization of a model of RV hypertrophy and failure, including RV-LV interaction, and to compare gene alterations between afterloaded RV versus LV. Pulmonary artery constriction was performed in 86 mice. Mice with mild and moderate pulmonary stenosis (PS) developed stable hypertrophy without decompensation. Mice with severe PS developed edema, decreased RV function, and high mortality. Tissue Doppler imaging demonstrated septal dyssynchrony and deleterious RV-LV interaction in the severe PS group. Microarray analysis showed 196 genes with increased expression and 1,114 with decreased expression. Several transcripts were differentially increased in the afterloaded RV but not in the afterloaded LV, including clusterin, neuroblastoma suppression of tumorigenicity 1, Dkk3, Sfrp2, formin binding protein, annexin A7, and lysyl oxidase. We have characterized a murine model of RV hypertrophy and failure, providing a platform for studying the physiological and molecular events of RV remodeling. Although the molecular responses of the RV and LV to afterload stress are mostly concordant, there are several key differences, which may represent targets for RV failure-specific therapy.
Collapse
Affiliation(s)
- Takashi Urashima
- Department of Pediatrics (Cardiology), Stanford University, Stanford, California, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Krijnen PAJ, Niessen HWM. The antiapoptotic protein clusterin protects cardiomyocytes against ischemia-induced cell death. Am J Physiol Heart Circ Physiol 2008; 293:H3223; author reply H3224. [PMID: 17989432 DOI: 10.1152/ajpheart.00964.2007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
46
|
Inhibition of NF-kappaB-dependent Bcl-xL expression by clusterin promotes albumin-induced tubular cell apoptosis. Kidney Int 2007; 73:567-77. [PMID: 18075502 DOI: 10.1038/sj.ki.5002563] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Apoptosis and inflammation, important contributors to the progression of chronic kidney disease, can be influenced by clusterin (a secreted glycoprotein that regulates apoptosis) and nuclear factor-kappaB (NF-kappaB, a transcription factor modifying the expression of inflammatory genes). We studied proteinuria-induced renal disease and its influence on clusterin-mediated apoptosis. Exposure of cultured mouse proximal tubule epithelial cells to bovine serum albumin (BSA) resulted in activation of NF-kappaB and activator protein-1 (AP-1) within hours followed by a decline in their activation, decreased activation of extracellular signal-regulated kinases (ERK1/2), decreased cell-associated antiapoptotic Bcl-xL protein but increased apoptosis. Clusterin progressively increased in the media over a 3 day period. Clusterin siRNA blocked protein production, increased NF-kappaB activation, and significantly increased cellular Bcl-xL protein, thereby reducing spontaneous and BSA-induced apoptosis. An siRNA to the NF-kappaB inhibitor IkappaBalpha had similar results. BSA-stimulated NF-kappaB activation reciprocally decreased AP-1 activity by preventing ERK1/2 phosphorylation. These in vitro studies suggest that clusterin inhibits NF-kappaB-mediated antiapoptotic effects by the apparent stabilization of IkappaBalpha switching from promoting inflammation to apoptosis during proteinuria.
Collapse
|
47
|
Sipkens JA, Krijnen PAJ, Meischl C, Cillessen SAGM, Smulders YM, Smith DEC, Giroth CPE, Spreeuwenberg MD, Musters RJP, Muller A, Jakobs C, Roos D, Stehouwer CDA, Rauwerda JA, van Hinsbergh VWM, Niessen HWM. Homocysteine affects cardiomyocyte viability: concentration-dependent effects on reversible flip-flop, apoptosis and necrosis. Apoptosis 2007; 12:1407-18. [PMID: 17440815 PMCID: PMC1914234 DOI: 10.1007/s10495-007-0077-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Hyperhomocysteinaemia (HHC) is thought to be a risk factor for cardiovascular disease including heart failure. While numerous studies have analyzed the role of homocysteine (Hcy) in the vasculature, only a few studies investigated the role of Hcy in the heart. Therefore we have analyzed the effects of Hcy on isolated cardiomyocytes. METHODS H9c2 cells (rat cardiomyoblast cells) and adult rat cardiomyocytes were incubated with Hcy and were analyzed for cell viability. Furthermore, we determined the effects of Hcy on intracellular mediators related to cell viability in cardiomyocytes, namely NOX2, reactive oxygen species (ROS), mitochondrial membrane potential (DeltaPsi (m)) and ATP concentrations. RESULTS We found that incubation of H9c2 cells with 0.1 mM D,L-Hcy (= 60 microM L-Hcy) resulted in an increase of DeltaPsi (m) as well as ATP concentrations. 1.1 mM D,L-Hcy (= 460 microM L-Hcy) induced reversible flip-flop of the plasma membrane phospholipids, but not apoptosis. Incubation with 2.73 mM D,L-Hcy (= 1.18 mM L-Hcy) induced apoptosis and necrosis. This loss of cell viability was accompanied by a thread-to-grain transition of the mitochondrial reticulum, ATP depletion and nuclear NOX2 expression coinciding with ROS production as evident from the presence of nitrotyrosin residues. Notably, only at this concentration we found a significant increase in S-adenosylhomocysteine which is considered the primary culprit in HHC. CONCLUSION We found concentration-dependent effects of Hcy in cardiomyocytes, varying from induction of reversible flip-flop of the plasma membrane phospholipids, to apoptosis and necrosis.
Collapse
Affiliation(s)
- Jessica A Sipkens
- Department of Pathology, VU University Medical Centre, Room 0E46, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Singla DK. Reply to “The antiapoptotic protein clusterin protects cardiomyocytes against ischemia-induced cell death”. Am J Physiol Heart Circ Physiol 2007. [DOI: 10.1152/ajpheart.01096.2007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Trougakos IP, Gonos ES. Regulation of clusterin/apolipoprotein J, a functional homologue to the small heat shock proteins, by oxidative stress in ageing and age-related diseases. Free Radic Res 2007; 40:1324-34. [PMID: 17090421 DOI: 10.1080/10715760600902310] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Clusterin/apolipoprotein J (CLU) gene has a nearly ubiquitous expression pattern in human tissues. The two main CLU protein isoforms in human cells include the conventional glycosylated secreted heterodimer (sCLU) and a truncated nuclear form (nCLU). CLU has been implicated in various physiological processes and in many severe physiological disturbance states including ageing, cancer progression, vascular damage, diabetes, kidney and neuron degeneration. Although unrelated in their etiology and clinical manifestation, these diseases represent states of increased oxidative stress, which in turn, promotes amorphous aggregation of target proteins, increased genomic instability and high rates of cellular death. Among the various properties attributed to CLU so far, those mostly investigated and invariably appreciated are its small heat shock proteins-like chaperone activity and its involvement in cell death regulation, which are both directly correlated to the main features of oxidant injury. Moreover, the presence of both a heat shock transcription factor-1 and an activator protein-1 element in the CLU gene promoter indicate that CLU gene can be an extremely sensitive biosensor to reactive oxygen species. This review emphasizes on CLU gene regulation by oxidative stress that is the common link between all pathological conditions where CLU has been implicated.
Collapse
Affiliation(s)
- Ioannis P Trougakos
- Laboratory of Molecular & Cellular Ageing, Institute of Biological Research & Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | | |
Collapse
|
50
|
Stejskal D, Lacnak B, Karpisek M, Kaminek M. OUR EXPERIENCES WITH MEASUREMENT OF NEW POTENTIAL BIOMARKERS IN THE DIAGNOSIS OF LATENT FORMS OF MYOCARDIAL ISCHEMIA. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2006; 150:239-44. [PMID: 17426785 DOI: 10.5507/bp.2006.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The need for a laboratory marker of myocardial ischemia has been alluded to for at least the last decade. AIM The aim of this study was to evaluate the diagnostic importance of the myosin light chain-1 (MLC-1), clusterin and Reg-Ialpha in patients with suspected myocardial ischemia. METHODS A group of 176 at high-risk for myocardial ischemia subjects was evaluated and divided into two subgroups using myocardial SPECT (Single Photon Emission Computed Tomography) - individuals with and without signs of myocardial ischemia. Laboratory markers in venous blood were repeatedly examined in all subjects: a) immediately prior to SPECT: C-reactive protein, Haemoglobin, Hematocrite, Lactate, MLC-1, Clusterin, Reg-Ialpha b) at subjective maximum: Hb, Htc, lactate, MLC-1, Clusterin, Reg-Ialpha c) 30 min after stress levels reached their peak: MLC-1, Clusterin, Reg-Ialpha and d) 60 min after peak stress levels: MLC-1, Clusterin, Reg-Ialpha. RESULTS Patients were divided into subgroups according to their positive and negative SPECT results (positive: n = 37; negative: n = 139). MLC-1 values were different for all 4 blood collections. An increase in MLC-1 > 2.2 mg/l showed 64 % sensitivity and 88 % specificity for the diagnosed presence of myocardial ischemia (AUC 0.81; LR+ 5.9; PPV+ 68 % and NPV- 87 %). There was no significant difference between the groups in terms of Clusterin and Reg-Ialpha for any of the sampling periods. CONCLUSIONS High diagnostic efficacy of detectable MLC-1 was shown for the diagnosis of latent myocardial ischemia. Measurement of serum Clusterin or Reg-Ialpha did not sufficient for the diagnosis of latent myocardial ischemia.
Collapse
Affiliation(s)
- David Stejskal
- Department of Laboratory Medicine and Internal Department, Sternberk Hospital, Czech Republic.
| | | | | | | |
Collapse
|