Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Biol Cell ; 113(1): 14-27, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32942336

ABSTRACT

BACKGROUND INFORMATION: Diabetes-induced testicular dysfunction is characterised by abnormal apoptosis of spermatogenic cells, but the underlying mechanism is poorly understood. This study aimed to investigate the roles of clusterin (CLU) in testicular damage associated with diabetes pathogenesis, as well as the molecular mechanism. A rat diabetes model was established using streptozocin, and the mouse spermatogenic cell line GC-1 spg was treated with high glucose as a cellular model. CLU was overexpressed in GC-1 spg cells, followed by detection of serum testosterone, cell proliferation, cell apoptosis and autophagy. RESULTS: CLU expression was significantly reduced and LC3 expression was elevated in testis tissues in the rat diabetes model and high glucose-treated GC-1 spg cells. High glucose led to suppressed viability, enhanced apoptosis, reduced Bcl-2 expression, elevated Bax expression and cleavage of Caspase-3/-9 in GC-1 spg cells, and these effects were abrogated by CLU overexpression. Additionally, CLU overexpression repressed LC3 and Beclin-1 expression, reduced the LC3II/LC3I ratio and promoted p62 expression in GC-1 spg cells in the presence of high glucose, and these effects were all mitigated by rapamycin treatment. Inhibition of PI3K/AKT/mTOR signalling with LY294002 activated autophagy in CLU-overexpressing GC-1 spg cells under high glucose conditions. CLU overexpression repressed autophagy and alleviated testicular damage in diabetic rats, which was also abrogated by LY294002 treatment. CONCLUSIONS: CLU expression is suppressed during diabetes-induced testicular damage, whereas CLU overexpression alleviates diabetes-induced testicular damage by activating PI3K/AKT/mTOR signalling to inhibit autophagy and further repress spermatogenic cell apoptosis.


Subject(s)
Clusterin/physiology , Diabetes Mellitus, Experimental/pathology , Testis , Animals , Apoptosis , Cell Line , Cell Proliferation , Male , Mice , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Testis/metabolism , Testis/pathology
2.
Reprod Biol Endocrinol ; 19(1): 56, 2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33874952

ABSTRACT

INTRODUCTION: Hypertension disorder of pregnancy (HDP) is one of the leading causes of maternal and foetal illness. The aim of the current study was to identify and verify novel serum markers for HDP. METHODS: A label-free LC-MS/MS method was used to establish the serum proteomic profiles of 12 pre-HDP (before clinical diagnosis of HDP) pregnancies and verify prioritized candidates in the verification set of 48 pre-HDP pregnancies. These biomarkers were revalidated by ELISA in an independent cohort of 88 pre-HDP pregnancies. Subsequently, the candidate biomarkers were histologically analysed by immunohistochemistry, and function was evaluated in TEV-1 cells. RESULTS: We identified 33 proteins with significantly increased abundance and 14 with decreased abundance (peptide FDR ≤ 1%, P < 0.05). Complement was one of the top enriched components in the pre-HDP group compared with the control group. Three complement factors (CLU, CFHR5, and CRP) were significantly increased in the three sets, of which CLU was a critical factor for the development of HDP (OR = 1.22, P < 0.001). When these three factors and body weight were combined, the AUC was 0.74, with a sensitivity of 0.67 and specificity of 0.68 for HDP prediction compared with normal pregnancy. In addition, inflammation-induced CLU could inhibit the invasion of TEV-1 cells. CONCLUSIONS: Complement proteins may play an essential role in the occurrence of HDP by acting on trophoblast cells. CLU may be a high-risk factor for HDP, and the models combining candidates show reasonable screening efficiency of HDP in the first half of pregnancy.


Subject(s)
Clusterin/physiology , Hypertension, Pregnancy-Induced/diagnosis , Maternal Serum Screening Tests/methods , Adult , Biomarkers/analysis , Biomarkers/blood , Blood Chemical Analysis/methods , Cells, Cultured , Chromatography, Liquid , Clusterin/blood , Cohort Studies , Complement System Proteins/analysis , Complement System Proteins/metabolism , Female , Humans , Hypertension, Pregnancy-Induced/blood , Predictive Value of Tests , Pregnancy , Pregnancy Trimester, First/blood , Pregnancy Trimester, Second/blood , Proteomics , Tandem Mass Spectrometry
3.
Neural Plast ; 2021: 9979157, 2021.
Article in English | MEDLINE | ID: mdl-34194490

ABSTRACT

Hearing loss is a debilitating disease that affects 10% of adults worldwide. Most sensorineural hearing loss is caused by the loss of mechanosensitive hair cells in the cochlea, often due to aging, noise, and ototoxic drugs. The identification of genes that can be targeted to slow aging and reduce the vulnerability of hair cells to insults is critical for the prevention of sensorineural hearing loss. Our previous cell-specific transcriptome analysis of adult cochlear hair cells and supporting cells showed that Clu, encoding a secreted chaperone that is involved in several basic biological events, such as cell death, tumor progression, and neurodegenerative disorders, is expressed in hair cells and supporting cells. We generated Clu-null mice (C57BL/6) to investigate its role in the organ of Corti, the sensory epithelium responsible for hearing in the mammalian cochlea. We showed that the deletion of Clu did not affect the development of hair cells and supporting cells; hair cells and supporting cells appeared normal at 1 month of age. Auditory function tests showed that Clu-null mice had hearing thresholds comparable to those of wild-type littermates before 3 months of age. Interestingly, Clu-null mice displayed less hair cell and hearing loss compared to their wildtype littermates after 3 months. Furthermore, the deletion of Clu is protected against aminoglycoside-induced hair cell loss in both in vivo and in vitro models. Our findings suggested that the inhibition of Clu expression could represent a potential therapeutic strategy for the alleviation of age-related and ototoxic drug-induced hearing loss.


Subject(s)
Clusterin/deficiency , Hair Cells, Auditory/physiology , Hearing Loss, Sensorineural/prevention & control , Presbycusis/prevention & control , Animals , Auditory Threshold , Base Sequence , CRISPR-Cas Systems , Cellular Senescence , Clusterin/biosynthesis , Clusterin/genetics , Clusterin/physiology , Drug Synergism , Evoked Potentials, Auditory, Brain Stem , Furosemide/administration & dosage , Furosemide/toxicity , Hair Cells, Auditory/drug effects , Hearing Loss, Sensorineural/chemically induced , Kanamycin/administration & dosage , Kanamycin/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Organ of Corti/pathology , Otoacoustic Emissions, Spontaneous , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
4.
Andrologia ; 52(2): e13498, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31833103

ABSTRACT

Recurrent pregnancy loss (RPL) is a perplexing problem experienced with two or more consecutive miscarriages wherein the cause remains unexplained in >50% of cases. However, despite several evidences of involvement of paternal factors on early embryogenesis and placental development, its contribution towards RPL has been largely unexplored. There is augmented lipid peroxidation, protein carbonylation, thionylation and enhanced histone retention in spermatozoa of RPL patients. Differentially expressed proteins in the spermatozoa of RPL patients may contribute towards aberrant embryo development and pregnancy loss. The present study comprised of male partners of RPL patients (n = 16) with the absence of any female factor abnormality and age-matched fertile healthy donors (n = 20). Pooled sperm samples from each group were subjected to high-throughput liquid chromatography-tandem mass spectrophotometry (LC-MS/MS) and subsequent bioinformatic analysis that identifies key proteins to be differentially expressed (DEPs). A total of 23 DEPs were identified with ≥2.0 fold change were considered to be significant. A key finding of the study was clusterin (CLUS), a predominant oxidative stress protein that takes part in an array of pre- and post-fertilisation molecular processes, found to be underexpressed as it was confirmed by Western blot analysis. This pilot study supports contributions of paternal oxidative predominance in RPL and encourages further investigation.


Subject(s)
Abortion, Habitual/etiology , Clusterin/metabolism , Proteomics , Abortion, Habitual/metabolism , Blotting, Western , Case-Control Studies , Chromatography, High Pressure Liquid , Clusterin/physiology , Female , Humans , Male , Pregnancy , Protein Interaction Maps , Proteins/analysis , Proteomics/methods , Semen/chemistry , Semen/metabolism , Spermatozoa/chemistry , Tandem Mass Spectrometry
5.
Exp Eye Res ; 147: 57-71, 2016 06.
Article in English | MEDLINE | ID: mdl-27131907

ABSTRACT

The multifunctional protein clusterin (CLU) was first described in 1983 as a secreted glycoprotein present in ram rete testis fluid that enhanced aggregation ('clustering') of a variety of cells in vitro. It was also independently discovered in a number of other systems. By the early 1990s, CLU was known under many names and its expression had been demonstrated throughout the body, including in the eye. Its homeostatic activities in proteostasis, cytoprotection, and anti-inflammation have been well documented, however its roles in health and disease are still not well understood. CLU is prominent at fluid-tissue interfaces, and in 1996 it was demonstrated to be the most highly expressed transcript in the human cornea, the protein product being localized to the apical layers of the mucosal epithelia of the cornea and conjunctiva. CLU protein is also present in human tears. Using a preclinical mouse model for desiccating stress that mimics human dry eye disease, the authors recently demonstrated that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration in the tears. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to LGALS3 (galectin-3), a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. CLU depletion from the ocular surface epithelia is seen in a variety of inflammatory conditions in humans and mice that lead to squamous metaplasia and a keratinized epithelium. This suggests that CLU might have a specific role in maintaining mucosal epithelial differentiation, an idea that can now be tested using the mouse model for desiccating stress. Most excitingly, the new findings suggest that CLU could serve as a novel biotherapeutic for dry eye disease.


Subject(s)
Clusterin/physiology , Cornea/metabolism , Animals , Clusterin/genetics , Clusterin/metabolism , Conjunctiva/metabolism , Disease Models, Animal , Dry Eye Syndromes/metabolism , Epithelium, Corneal/metabolism , Humans , Inflammation/metabolism , Tears/metabolism
6.
Lipids Health Dis ; 14: 114, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26391229

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/gp91(phox) 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.


Subject(s)
Angiotensin II/physiology , Clusterin/physiology , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Cells, Cultured , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protective Factors , Rats, Sprague-Dawley , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 109(17): 6745-50, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22493272

ABSTRACT

The ability to sense temperature is essential for organism survival and efficient metabolism. Body temperatures profoundly affect many physiological functions, including immunity. Transient receptor potential melastatin 2 (TRPM2) is a thermosensitive, Ca(2+)-permeable cation channel expressed in a wide range of immunocytes. TRPM2 is activated by adenosine diphosphate ribose and hydrogen peroxide (H(2)O(2)), although the activation mechanism by H(2)O(2) is not well understood. Here we report a unique activation mechanism in which H(2)O(2) lowers the temperature threshold for TRPM2 activation, termed "sensitization," through Met oxidation and adenosine diphosphate ribose production. This sensitization is completely abolished by a single mutation at Met-214, indicating that the temperature threshold of TRPM2 activation is regulated by redox signals that enable channel activity at physiological body temperatures. Loss of TRPM2 attenuates zymosan-evoked macrophage functions, including cytokine release and fever-enhanced phagocytic activity. These findings suggest that redox signals sensitize TRPM2 downstream of NADPH oxidase activity and make TRPM2 active at physiological body temperature, leading to increased cytosolic Ca(2+) concentrations. Our results suggest that TRPM2 sensitization plays important roles in macrophage functions.


Subject(s)
Clusterin/physiology , Macrophages/physiology , Cell Line , Humans , Oxidation-Reduction , Temperature
8.
Neuroendocrinology ; 98(3): 233-41, 2013.
Article in English | MEDLINE | ID: mdl-24080898

ABSTRACT

Clusterin is a sulfated glycoprotein abundantly expressed in the pituitary gland and hypothalamus of mammals. However, its physiological role in neuroendocrine function is largely unknown. In the present study, we investigated the effects of intracerebroventricular (ICV) administration of clusterin on plasma pituitary hormone levels in normal rats. Single ICV injection of clusterin provoked neurohormonal changes seen under acute stress condition: increased plasma adrenocorticotropic hormone (ACTH), corticosterone, GH and prolactin levels and decreased LH and FSH levels. Consistently, hypothalamic and pituitary clusterin expression levels were upregulated following a restraint stress, suggesting an involvement of endogenous clusterin in stress-induced neurohormonal changes. In the pituitary intermediate lobe, clusterin was coexpressed with proopiomelanocortin (POMC), a precursor of ACTH. Treatment of clusterin in POMC expressing AtT-20 pituitary cells increased basal and corticotropin-releasing hormone (CRH)-stimulated POMC promoter activities and intracellular cAMP levels. Furthermore, clusterin treatment triggered ACTH secretion from AtT-20 cells in a CRH-dependent manner, indicating that increased clusterin under stressful conditions may augment CRH-stimulated ACTH production and release. In summary, hypothalamic and pituitary clusterin may function as a modulator of neurohormonal responses under stressful conditions.


Subject(s)
Clusterin/physiology , Hypothalamus/metabolism , Neurotransmitter Agents/biosynthesis , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/biosynthesis , Adrenocorticotropic Hormone/metabolism , Animals , Clusterin/administration & dosage , Clusterin/blood , Hypothalamus/drug effects , Injections, Intraventricular , Male , Neurotransmitter Agents/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Pituitary Gland/drug effects , Pro-Opiomelanocortin/antagonists & inhibitors , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/blood , Stress, Psychological/prevention & control , Stress, Psychological/psychology , Up-Regulation/physiology
9.
Hepatogastroenterology ; 60(121): 70-5, 2013.
Article in English | MEDLINE | ID: mdl-22975582

ABSTRACT

BACKGROUND/AIMS: Non-small cell lung cancer (NSCLC) constitutes around 85% of lung cancer cases and is frequently beyond surgical intervention. METHODOLOGY: Secretory clusterin (sCLU) is found in diverse types of human cancers and is unregulated in a variety of cell lines in response to stress, and enhances cancer cell survival. However, the roles of sCLU in NSCLC are still to be elucidated. RESULTS: Here we show that RNA interference (RNAi)-mediated sCLU gene silencing with short interference RNA (siRNA) strongly decreased the sCLU mRNA and protein levels, as well as suppressed cell proliferation and induced cell apoptosis. In addition, sCLU siRNA also blocked the PI3K/AKT signaling pathway, and decreased the AKT phosphorylation level, but no change was found in total AKT level. More importantly, PI3K/AKT signaling pathway inhibitor, LY294002, also reduced tumor cell proliferation, which is similar to the result with or without sCLU siRNA treatment. CONCLUSIONS: These results suggest that sCLU plays a positive role in NSCLC cell proliferation, which may be mediated by the PI3K/AKT signaling pathway. Our work in this study demonstrates RNAi-mediated sCLU gene silencing may provide a novel therapeutic strategy in the treatment of NSCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Clusterin/genetics , Lung Neoplasms/therapy , RNA Interference , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Clusterin/physiology , Humans , Lung Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology
10.
J Cell Physiol ; 227(3): 1157-67, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21567405

ABSTRACT

Clusterin (CLU), a glycoprotein, is involved in apoptosis, producing two alternatively spliced isoforms in various cell types. The pro-apoptotic CLU appears to be a nuclear isoform (nuclear clusterin; nCLU), and the secretory CLU (sCLU) is thought to be anti-apoptotic. The detailed molecular mechanism of nCLU as a pro-apoptotic molecule has not yet been clear. In the current study, overexpressed nCLU induced apoptosis in human kidney cells. Biochemical studies revealed that nCLU sequestered Bcl-XL via a putative BH3 motif in the C-terminal coiled coil (CC2) domain, releasing Bax, and promoted apoptosis accompanied by activation of caspase-3 and cytochrome c release. These results suggest a novel mechanism of apoptosis mediated by nCLU as a pro-apoptotic molecule.


Subject(s)
Apoptosis , Cell Survival , Clusterin/physiology , bcl-X Protein/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/genetics , Clusterin/chemistry , Clusterin/genetics , HEK293 Cells , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Male , Molecular Sequence Data , Prostatic Neoplasms , Protein Interaction Domains and Motifs/genetics , Protein Structure, Tertiary/physiology , bcl-X Protein/chemistry , bcl-X Protein/genetics
11.
Biochem Biophys Res Commun ; 420(4): 851-6, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22465014

ABSTRACT

Clusterin is a disulfide-linked heterodimeric glycoprotein that has been implicated in a variety of biological processes. Its expression has been shown to be elevated during cellular senescence and normal aging, but it is uncertain whether clusterin protects against aging or whether its expression is a consequence of aging. To investigate the functions of clusterin during organismal aging, we established transgenic Drosophila alleles to induce the expression of the secretory form of human clusterin (hClu(S)) using the Gal4/UAS system. hClu(S) protein (~60 kDa) was detected in both adult homogenates and larval hemolymphs of flies ubiquitously overexpressing hClu(S) (da-Gal4>UAS-hClu(S)) and in motoneurons (D42-Gal4>UAS-hClu(S)). Interestingly, the mean lifespans of these hClu(S)-overexpressing flies were significantly greater than those of control flies that exhibited no hClu(S) induction. hClu(S)-overexpressing flies also showed significantly greater tolerance to heat shock, wet starvation, and oxidative stress. Furthermore, amounts of reactive oxygen species (ROS) in whole bodies were significantly lower in hClu(S)-overexpressing flies. In addition, clusterin was found to prevent the inactivation of glutamine synthetase (GS) by metal-catalyzed oxidation (MCO) in vitro, and this protection was only supported by thiol-reducing equivalents, such as, DTT or GSH, and not by ascorbate (a non-thiol MCO system). Furthermore, this protection against GS inactivation by clusterin was abolished by reacting clusterin with N-ethylmaleimide, a sulfhydryl group-modifying agent. Taken together, these results suggest that a disulfide-linked form of clusterin functions as an antioxidant protein via its cysteine sulfhydryl groups to reduce ROS levels and delay the organismal aging in fruit flies.


Subject(s)
Clusterin/physiology , Drosophila melanogaster/physiology , Heat-Shock Response/genetics , Longevity/genetics , Oxidative Stress/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/physiology , Clusterin/genetics , Dithiothreitol/pharmacology , Drosophila Proteins , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Ethylmaleimide/pharmacology , Glutamate-Ammonia Ligase , Hemolymph/metabolism , Humans , Longevity/drug effects , Reactive Oxygen Species/metabolism
12.
Biochem Biophys Res Commun ; 422(1): 200-5, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22575505

ABSTRACT

Tumor associated macrophages are known to be closely linked with tumor progression and metastasis. On the other hand, clusterin is overexpressed in several tumor types and regarded as a putative tumor-promoting factor due to this overexpression and the subsequent induction of chemoresistance. In our previous study, clusterin was found to induce the expression of matrix metalloproteinase-9 (MMP-9) in macrophages, and MMP-9 is known to be essential for tumor cell migration and invasion via basement membrane breakdown. Because paracrine interactions between tumor cells and surrounding macrophages regulate metastasis, these findings raise the possibility that clusterin promotes the secretion of cytokines in macrophages in addition to MMP-9. Here, we demonstrate that clusterin upregulates the expressions of chemotactic cytokines, that is, monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1ß (MIP-1ß), regulated upon activation, normal T cell expressed and secreted (RANTES), and tumor necrosis factor-α (TNF-α) in Raw264.7 macrophages. In particular, clusterin stimulated TNF-α secretion via the activations of ERK, JNK, and PI3K/Akt pathways in a time and dose-dependent manner. Furthermore, clusterin-induced TNF-α secretion was found to play a critical role in the chemotactic migration of Raw264.7 macrophages. It was also found that clusterin acts directly as a chemoattractant for macrophages. Together, these results suggest that clusterin stimulates the expression and secretion of TNF-α, which plays a critical role in promoting macrophage chemotaxis, via ERK, JNK, and PI3K/Akt pathways. Collectively, these findings describe a novel function for clusterin as an inducer of TNF-α in macrophages and their chemotactic migration, and suggest that clusterin has a tumor-promoting effect.


Subject(s)
Chemotaxis/physiology , Clusterin/physiology , Macrophages, Peritoneal/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Chemotaxis/drug effects , Clusterin/pharmacology , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Signaling System , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
13.
Int J Gynecol Cancer ; 22(3): 465-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21633299

ABSTRACT

OBJECTIVE: Overexpression of clusterin (CLU), an antiapoptotic molecule, has been reported to induce resistance to radiotherapy (RT) in a variety of cancer cell types. The aim of this study was to evaluate the significance of CLU expression to predict survival of patients with advanced-stage cervical cancer who received curative intended RT. METHODS: Biopsy tissue specimens of advanced-stage cervical cancer before curative intended RT were obtained from 34 patients who were treated at Hokkaido University Hospital between 1998 and 2008 and whose complete medical records were available. The expression of CLU protein was analyzed by immunohistochemistry. Findings were evaluated in relation to several clinicopathological factors. Survival analyses were performed using the Kaplan-Meier curves and the log-rank test. Independent prognostic factors were determined by multivariate Cox regression analysis. RESULTS: Clusterin protein was mainly present in the cytoplasm of cervical cancer cells. The expression of CLU protein in cervical cancer tissues before curative intended RT was not significantly related to any clinicopathological factors analyzed, including age, clinical stage, histologic type, and response to RT. Univariate analysis on prognostic factors showed that histologic type (P = 0.001), and CLU expression (P = 0.02) were related to survival. Multivariate analysis revealed that both histologic type (P = 0.002), and CLU expression (P = 0.02) were independent prognostic factors for overall survival. CONCLUSION: We conclude that CLU could be a new molecular marker to predict overall survival of patients with advanced-stage cervical cancer treated with curative intended RT.


Subject(s)
Carcinoma/radiotherapy , Clusterin/metabolism , Uterine Cervical Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Carcinoma/metabolism , Carcinoma/pathology , Clusterin/physiology , Disease Progression , Female , Humans , Immunohistochemistry , Intention , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Prognosis , Radiation Oncology/methods , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
14.
Tumour Biol ; 32(2): 285-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21042904

ABSTRACT

Clusterin is a ubiquitously expressed glycoprotein with multiple binding partners including IL-6, Ku70, and Bax. Clusterin blocks apoptosis by binding to activated Bax and sequestering it in the cytoplasm, thereby preventing Bax from entering mitochondria, releasing cytochrome c, and triggering apoptosis. Because increased clusterin expression correlates with aggressive behavior in tumors, clusterin inhibition might be beneficial in cancer treatment. Our recent findings indicated that, in neuroblastoma cells, cytoplasmic Bax also binds to Ku70; when Ku70 is acetylated, Bax is released and can initiate cell death. Therefore, increasing Ku70 acetylation, such as by using histone deacetylase inhibitors, may be therapeutically useful in promoting cell death in neuroblastoma tumors. Since clusterin, Bax, and Ku70 form a complex, it seemed likely that clusterin would mediate its anti-apoptotic effects by inhibiting Ku70 acetylation and blocking Bax release. Our results, however, demonstrate that while clusterin level does indeed determine the sensitivity of neuroblastoma cells to histone deacetylase inhibitor-induced cell death, it does so without affecting histone deacetylase-inhibitor-induced Ku70 acetylation. Our results suggest that in neuroblastoma, clusterin exerts its anti-apoptotic effects downstream of Ku70 acetylation, likely by directly blocking Bax activation.


Subject(s)
Apoptosis/drug effects , Clusterin/physiology , Histone Deacetylase Inhibitors/pharmacology , Neuroblastoma/pathology , Acetylation/drug effects , Antigens, Nuclear/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/physiology , Cell Line, Tumor , Cell Survival/drug effects , Clusterin/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Humans , Ku Autoantigen , Neuroblastoma/physiopathology , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/physiology
15.
Neurotox Res ; 39(6): 2108-2119, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32820456

ABSTRACT

Alzheimer's disease (AD) is a chronic and devastating neurodegenerative disorder that is affecting elderly people at an increasing rate. Clusterin (CLU), an extracellular chaperone, is an ubiquitously expressed protein that can be identified in various body fluids and tissues. Expression of CLU can lead to various processes including suppression of complement system, lipid transport, chaperone function, and also controlling neuronal cell death and cell survival mechanisms. Studies have confirmed that the level of CLU expression is increased in AD. Furthermore, CLU also decreased the toxicity and aggregation of amyloid beta (Aß). However when the Aß level was far greater than CLU, then the amyloid generation was increased. CLU was also found to incorporate in the amyloid aggregates, which were more harmful as compared with the Aß42 aggregates alone. Growing evidence indicates that CLU plays roles in AD pathogenesis via various processes, including aggregation and clearance of Aß, neuroinflammation, lipid metabolism, Wnt signaling, copper homeostasis, and regulation of neuronal cell cycle and apoptosis. In this article, we represent the critical interaction of CLU and AD based on recent advances. Furthermore, we have also focused on the Aß-dependent and Aß-independent mechanisms by which CLU plays a role in AD pathogenesis.


Subject(s)
Alzheimer Disease/etiology , Clusterin/physiology , Animals , Clusterin/metabolism , Humans
16.
Commun Biol ; 4(1): 564, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980978

ABSTRACT

The risks of non-alcoholic fatty liver disease (NAFLD) include obese and non-obese stresses such as chronic hepatitis C virus (HCV) infection, but the regulatory determinants remain obscure. Apolipoprotein J (ApoJ) served as an ER-Golgi contact-site chaperone near lipid droplet (LD), facilitating HCV virion production. We hypothesized an interplay between hepatic ApoJ, cholesterol esterification and lipid deposit in response to NAFLD inducers. Exposures of HCV or free-fatty acids exhibited excess LDs along with increased ApoJ expression, whereas ApoJ silencing alleviated hepatic lipid accumulation. Both stresses could concomitantly disperse Golgi, induce closer ApoJ and sterol O-acyltransferase 2 (SOAT2) contacts via the N-terminal intrinsically disordered regions, and increase cholesteryl-ester. Furthermore, serum ApoJ correlated positively with cholesterol and low-density lipoprotein levels in normal glycaemic HCV patients, NAFLD patients and in mice with steatosis. Taken together, hepatic ApoJ might activate SOAT2 to supply cholesteryl-ester for lipid loads, thus providing a therapeutic target of stress-induced steatosis.


Subject(s)
Clusterin/metabolism , Lipid Metabolism/physiology , Sterol O-Acyltransferase/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cholesterol/metabolism , Cholesterol Esters/metabolism , Clusterin/physiology , Esterification , Fatty Liver/metabolism , Female , Hepatitis C, Chronic/metabolism , Humans , Hypercholesterolemia/metabolism , Lipid Droplets/metabolism , Lipids/physiology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/physiopathology , Sterol O-Acyltransferase/physiology , Sterol O-Acyltransferase 2
17.
Arterioscler Thromb Vasc Biol ; 29(10): 1558-64, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19696405

ABSTRACT

OBJECTIVE: Clusterin is induced in vascular smooth muscle cells (VSMCs) during atherosclerosis and injury-induced neointimal hyperplasia. However, its functional roles in VSMCs and endothelial cells remain controversial and elusive. This study was undertaken to clarify the role of clusterin in neointimal hyperplasia and elucidate its mechanism of action. METHODS AND RESULTS: Adenovirus-mediated overexpression of clusterin (Ad-Clu) repressed TNF-alpha-stimulated expression of MCP-1, fractalkine, ICAM-1, VCAM-1, and MMP-9, leading to inhibition of VSMC migration. Both Ad-Clu and secreted clusterin suppressed VSMC proliferation by inhibiting DNA synthesis, but not by inducing apoptosis. Ad-Clu upregulated p53 and p21(cip1/waf1) but downregulated cyclins D and E, leading to suppression of pRb phosphorylation and subsequent induction of G1 arrest in VSMCs. Clusterin deficiency augmented VSMC proliferation in vitro and accelerated neointimal hyperplasia in vivo, but concomitantly impaired reendothelialization in wire-injured murine femoral arteries. Moreover, Ad-Clu significantly reduced neointimal thickening in balloon-injured rat carotid arteries. Clusterin also diminished TNF-alpha-induced apoptosis of human umbilical vein endothelial cells and restored endothelial nitric oxide synthase expression suppressed by TNF-alpha. CONCLUSIONS: These results suggest that upregulation of clusterin during vascular injury may be a protective response against, rather than a causative response to, the development of neointimal hyperplasia.


Subject(s)
Clusterin/physiology , Cytoprotection , Endothelial Cells/cytology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Tunica Intima/pathology , Animals , Cell Movement , Cell Proliferation , DNA/biosynthesis , G1 Phase , Hyperplasia , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Phosphorylation , Rats , Rats, Sprague-Dawley , Retinoblastoma Protein/metabolism
18.
Clin Cancer Res ; 15(1): 48-59, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118032

ABSTRACT

PURPOSE: Secretory clusterin (sCLU)/apolipoprotein J is an extracellular chaperone that has been functionally implicated in DNA repair, cell cycle regulation, apoptotic cell death, and tumorigenesis. It exerts a prosurvival function against most therapeutic treatments for cancer and is currently an antisense target in clinical trials for tumor therapy. However, the molecular mechanisms underlying its function remained largely unknown. EXPERIMENTAL DESIGN: The molecular effects of small interfering RNA-mediated sCLU depletion in nonstressed human cancer cells were examined by focusing entirely on the endogenously expressed sCLU protein molecules and combining molecular, biochemical, and microscopic approaches. RESULTS: We report here that sCLU depletion in nonstressed human cancer cells signals stress that induces p53-dependent growth retardation and high rates of endogenous apoptosis. We discovered that increased apoptosis in sCLU-depleted cells correlates to altered ratios of proapoptotic to antiapoptotic Bcl-2 protein family members, is amplified by p53, and is executed by mitochondrial dysfunction. sCLU depletion-related stress signals originate from several sites, because sCLU is an integral component of not only the secretory pathway but also the nucleocytosolic continuum and mitochondria. In the cytoplasm, sCLU depletion disrupts the Ku70-Bax complex and triggers Bax activation and relocation to mitochondria. We show that sCLU binds and thereby stabilizes the Ku70-Bax protein complex serving as a cytosol retention factor for Bax. CONCLUSIONS: We suggest that elevated sCLU levels may enhance tumorigenesis by interfering with Bax proapoptotic activities and contribute to one of the major characteristics of cancer cells, that is, resistance to apoptosis.


Subject(s)
Antigens, Nuclear/metabolism , Clusterin/physiology , DNA-Binding Proteins/metabolism , Genes, p53 , Mitochondria/physiology , bcl-2-Associated X Protein/metabolism , Cell Line, Tumor , Cytosol/metabolism , Humans , Ku Autoantigen , Organelles/metabolism , RNA, Small Interfering/pharmacology
19.
Neoplasma ; 57(3): 191-7, 2010.
Article in English | MEDLINE | ID: mdl-20353268

ABSTRACT

Clusterin (CLU) is expressed in a wide variety of human tissues and fluids. Overexpression of cytoplasmic clusterin (sCLU) has been implicated in cancer development and progression. The aim of the present study is to evaluate the association of sCLU overexpression with clinicopathological features of human gastric carcinomas (GC).We constructed a gastric cancer tissue microarray containing 173 primary gastric carcinomas and 70 paired non-neoplastic mucosa specimens. The expression of sCLU was studied by immunohistochemistry. The correlations between sCLU expression and clinicopathological features, p53 abnormality, as well as Ki67 activation were analyzed. Overexpressions of sCLU was detected in 28.5% (n=165) of primary GCs by immunohistochemical staining, but not in non-neoplastic mucosa. Clinical association study found that overexpression of sCLU was significantly correlated with lymph-node metastasis (p < 0.001), tumor invasion (p < 0.001) and TNM stage (p < 0.001). In Kaplan-Meier survival analysis, overexpression of sCLU was significantly correlated with unfavorable survival in advanced GCs (p < 0.03). Furthermore, the association of sCLU with abnormal expression of p53 was ascertained. These results suggested that overexpression of sCLU was involved in the progression of GC and it's oncogenic function might be associated with p53 abnormality. Overexpression of sCLU seems to be related with patient's shorter survival in late stage GC.


Subject(s)
Clusterin/physiology , Stomach Neoplasms/pathology , Tissue Array Analysis/methods , Adult , Aged , Aged, 80 and over , Clusterin/analysis , Cytoplasm/chemistry , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Stomach Neoplasms/chemistry , Stomach Neoplasms/mortality , Tumor Suppressor Protein p53/analysis
20.
Sci Rep ; 10(1): 14888, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913257

ABSTRACT

Clusterin, a glycoprotein encoded by the CLU gene, is expressed in many tissues, including the kidney, and clusterin expression is upregulated in the glomeruli of patients with various forms of kidney disease. Here, we investigated the role of clusterin in diabetic nephropathy (DN). In this study, we found that glomerular clusterin expression was increased in both patients with DN and streptozotocin-induced diabetic mice and that it co-localised with the podocyte marker WT1, indicating clusterin is expressed in podocytes. In our in vitro analysis, we found no significant change in CLU mRNA expression in podocytes following stimulation with high glucose and angiotensin II; in contrast, CLU mRNA expression was significantly upregulated following methylglyoxal stimulation. Methylglyoxal treatment also significantly decreased the mRNA expression of the slit diaphragm markers ZO-1 and NEPH1 and significantly increased the mRNA expression of the oxidative stress marker HO-1. Lastly, we showed that pre-incubating podocytes with recombinant human clusterin protein increased podocyte survival, prevented slit diaphragm damage, and reduced oxidative stress‒induced apoptosis following methylglyoxal stimulation. Taken together, our results indicate that glomerular clusterin is upregulated in DN, and this increase in clusterin expression may protect against oxidative stress-induced apoptosis in podocytes, providing a possible new therapeutic target for DN and other kidney diseases.


Subject(s)
Apoptosis/physiology , Clusterin/metabolism , Diabetic Nephropathies/metabolism , Kidney Glomerulus/metabolism , Oxidative Stress/physiology , Podocytes/cytology , Clusterin/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL