51
|
Kubis AM, Piwowar A. The new insight on the regulatory role of the vitamin D3 in metabolic pathways characteristic for cancerogenesis and neurodegenerative diseases. Ageing Res Rev 2015; 24:126-37. [PMID: 26238411 DOI: 10.1016/j.arr.2015.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 07/27/2015] [Indexed: 12/14/2022]
Abstract
Apart from the classical function of regulating intestinal, bone and kidney calcium and phosphorus absorption as well as bone mineralization, there is growing evidence for the neuroprotective function of vitamin D3 through neuronal calcium regulation, the antioxidative pathway, immunomodulation and detoxification. Vitamin D3 and its derivates influence directly or indirectly almost all metabolic processes such as proliferation, differentiation, apoptosis, inflammatory processes and mutagenesis. Such multifactorial effects of vitamin D3 can be a profitable source of new therapeutic solutions for two radically divergent diseases, cancer and neurodegeneration. Interestingly, an unusual association seems to exist between the occurrence of these two pathological states, called "inverse comorbidity". Patients with cognitive dysfunctions or dementia have considerably lower risk of cancer, whereas survivors of cancer have lower prevalence of central nervous system (CNS) disorders. To our knowledge, there are few publications analyzing the role of vitamin D3 in biological pathways existing in carcinogenic and neuropathological disorders.
Collapse
Affiliation(s)
- Adriana Maria Kubis
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 Str., 50-552 Wrocław, Poland.
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 Str., 50-552 Wrocław, Poland
| |
Collapse
|
52
|
Song B, Bian Q, Zhang YJ, Shao CH, Li G, Liu AA, Jing W, Liu R, Zhou YQ, Jin G, Hu XG. Downregulation of ASPP2 in pancreatic cancer cells contributes to increased resistance to gemcitabine through autophagy activation. Mol Cancer 2015; 14:177. [PMID: 26438046 PMCID: PMC4594892 DOI: 10.1186/s12943-015-0447-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/21/2015] [Indexed: 01/05/2023] Open
Abstract
Background Apoptosis-stimulating of p53 protein 2 (ASPP2) is one of the ASPP family members and it has been reported to be associated with human cancer. However, the role of it in pancreatic cancer is still not clear. Methods We analyzed the expression level of ASPP2 in cancer tissue samples with RT-qPCR, Western Blotting assay and immunohistochemistry staining. We studied the biological function of ASPP2 and its mechanism with gene overexpression and gene silencing technologies. We determined the sensitivity of pancreatic cells with differential ASPP2 level to gemcitabine and whether autophagy inhibition affected the gemcitabine resistance, both in vitro and in vivo. Results Expression of ASPP2 was downregulated in cancerous tissues in comparison with para-cancerous tissues. ASPP2 expression was linked to clinical outcomes in patients and down-regulation of ASPP2 increased cell proliferation, autophagic flux, the activity of AMP Kinase of pancreatic cancer cells and vice versa. Knockdown of ASPP2 results in increased resistance to gemcitabine, which was attributed to the enhanced autophagy. Conclusions ASSP2 expression is lower in cancerous tissues and decreased ASPP2 lead to higher cancer cells proliferation and autophagic flux, which contribute to the gemcitabine resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0447-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bin Song
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Qi Bian
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| | - Yi-Jie Zhang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Cheng-Hao Shao
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Gang Li
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - An-An Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Wei Jing
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Rui Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Ying-Qi Zhou
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Gang Jin
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China. .,Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| | - Xian-Gui Hu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China. .,Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| |
Collapse
|
53
|
Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and Prevents Malignant Transformation. Cell Stem Cell 2015; 17:23-34. [DOI: 10.1016/j.stem.2015.05.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 04/02/2015] [Accepted: 05/26/2015] [Indexed: 12/23/2022]
|
54
|
A Screen for Extracellular Signal-Regulated Kinase-Primed Glycogen Synthase Kinase 3 Substrates Identifies the p53 Inhibitor iASPP. J Virol 2015; 89:9232-41. [PMID: 26109723 DOI: 10.1128/jvi.01072-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/19/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The Kaposi's sarcoma-associated herpesvirus (KSHV) LANA protein is essential for the replication and maintenance of virus genomes in latently KSHV-infected cells. LANA also drives dysregulated cell growth through a multiplicity of mechanisms that include altering the activity of the cellular kinases extracellular signal-regulated kinase (ERK) and glycogen synthase kinase 3 (GSK-3). To investigate the potential impact of these changes in enzyme activity, we used protein microarrays to identify cell proteins that were phosphorylated by the combination of ERK and GSK-3. The assays identified 58 potential ERK-primed GSK-3 substrates, of which 23 had evidence for in vivo phosphorylation in mass spectrometry databases. Two of these, SMAD4 and iASPP, were selected for further analysis and were confirmed as ERK-primed GSK-3 substrates. Cotransfection experiments revealed that iASPP, but not SMAD4, was targeted for degradation in the presence of GSK-3. iASPP interferes with apoptosis induced by p53 family members. To determine the importance of iASPP to KSHV-infected-cell growth, primary effusion lymphoma (PEL) cells were treated with an iASPP inhibitor in the presence or absence of the MDM2 inhibitor Nutlin-3. Drug inhibition of iASPP activity induced apoptosis in BC3 and BCBL1 PEL cells but did not induce poly(ADP-ribose) polymerase (PARP) cleavage in virus-negative BJAB cells. The effect of iASPP inhibition was additive with that of Nutlin-3. Interfering with iASPP function is therefore another mechanism that can sensitize KSHV-positive PEL cells to cell death. IMPORTANCE KSHV is associated with several malignancies, including primary effusion lymphoma (PEL). The KSHV-encoded LANA protein is multifunctional and promotes both cell growth and resistance to cell death. LANA is known to activate ERK and limit the activity of another kinase, GSK-3. To discover ways in which LANA manipulation of these two kinases might impact PEL cell survival, we screened a human protein microarray for ERK-primed GSK-3 substrates. One of the proteins identified, iASPP, showed reduced levels in the presence of GSK-3. Further, blocking iASPP activity increased cell death, particularly in p53 wild-type BC3 PEL cells.
Collapse
|
55
|
iASPP, a previously unidentified regulator of desmosomes, prevents arrhythmogenic right ventricular cardiomyopathy (ARVC)-induced sudden death. Proc Natl Acad Sci U S A 2015; 112:E973-81. [PMID: 25691752 DOI: 10.1073/pnas.1408111112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Desmosomes are anchoring junctions that exist in cells that endure physical stress such as cardiac myocytes. The importance of desmosomes in maintaining the homeostasis of the myocardium is underscored by frequent mutations of desmosome components found in human patients and animal models. Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a phenotype caused by mutations in desmosomal components in ∼ 50% of patients, however, the causes in the remaining 50% of patients still remain unknown. A deficiency of inhibitor of apoptosis-stimulating protein of p53 (iASPP), an evolutionarily conserved inhibitor of p53, caused by spontaneous mutation recently has been associated with a lethal autosomal recessive cardiomyopathy in Poll Hereford calves and Wa3 mice. However, the molecular mechanisms that mediate this putative function of iASPP are completely unknown. Here, we show that iASPP is expressed at intercalated discs in human and mouse postmitotic cardiomyocytes. iASPP interacts with desmoplakin and desmin in cardiomyocytes to maintain the integrity of desmosomes and intermediate filament networks in vitro and in vivo. iASPP deficiency specifically induces right ventricular dilatation in mouse embryos at embryonic day 16.5. iASPP-deficient mice with exon 8 deletion (Ppp1r13l(Δ8/Δ8)) die of sudden cardiac death, displaying features of ARVC. Intercalated discs in cardiomyocytes from four of six human ARVC cases show reduced or loss of iASPP. ARVC-derived desmoplakin mutants DSP-1-V30M and DSP-1-S299R exhibit weaker binding to iASPP. These data demonstrate that by interacting with desmoplakin and desmin, iASPP is an important regulator of desmosomal function both in vitro and in vivo. This newly identified property of iASPP may provide new molecular insight into the pathogenesis of ARVC.
Collapse
|
56
|
Kramer D, Schön M, Bayerlová M, Bleckmann A, Schön MP, Zörnig M, Dobbelstein M. A pro-apoptotic function of iASPP by stabilizing p300 and CBP through inhibition of BRMS1 E3 ubiquitin ligase activity. Cell Death Dis 2015; 6:e1634. [PMID: 25675294 PMCID: PMC4669821 DOI: 10.1038/cddis.2015.17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/27/2014] [Accepted: 01/02/2015] [Indexed: 12/26/2022]
Abstract
The p53 family and its cofactors are potent inducers of apoptosis and form a barrier to cancer. Here, we investigated the impact of the supposedly inhibitory member of the apoptosis-stimulating protein of p53, iASPP, on the activity of the p53 homolog TAp73, and its cofactors p300 and CBP. We found that iASPP interacted with and stabilized the histone acetyltransferase p300 and its homolog CBP upon cisplatin treatment. Vice versa, iASPP depletion by shRNA resulted in decreased amounts of p300 and CBP, impaired binding of p300 and TAp73 to target site promoters, reduced induction of pro-apoptotic TAp73 target genes, and impaired apoptosis. Mechanistically, we observed that the p300-regulatory E3 ubiquitin ligase BRMS1 could rescue the degradation of p300 and CBP in cisplatin-treated, iASPP-depleted cells. This argues that iASPP stabilizes p300 and CBP by interfering with their BRMS1-mediated ubiquitination, thereby contributing to apoptotic susceptibility. In line, iASPP overexpression partially abolished the interaction of BRMS1 and CBP upon DNA damage. Reduced levels of iASPP mRNA and protein as well as CBP protein were observed in human melanoma compared with normal skin tissue and benign melanocytic nevi. In line with our findings, iASPP overexpression or knockdown of BRMS1 each augmented p300/CBP levels in melanoma cell lines, thereby enhancing apoptosis upon DNA damage. Taken together, destabilization of p300/CBP by downregulation of iASPP expression levels appears to represent a molecular mechanism that contributes to chemoresistance in melanoma cells.
Collapse
Affiliation(s)
- D Kramer
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - M Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - M Bayerlová
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - A Bleckmann
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - M P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - M Zörnig
- Institute of Tumor Biology and Experimental Therapy, Georg Speyer Haus, Frankfurt am Main,Germany
| | - M Dobbelstein
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
57
|
Yoh K, Prywes R. Pathway Regulation of p63, a Director of Epithelial Cell Fate. Front Endocrinol (Lausanne) 2015; 6:51. [PMID: 25972840 PMCID: PMC4412127 DOI: 10.3389/fendo.2015.00051] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/02/2015] [Indexed: 02/03/2023] Open
Abstract
The p53-related gene p63 is required for epithelial cell establishment and its expression is often altered in tumor cells. Great strides have been made in understanding the pathways and mechanisms that regulate p63 levels, such as the Wnt, Hedgehog, Notch, and EGFR pathways. We discuss here the multiple signaling pathways that control p63 expression as well as transcription factors and post-transcriptional mechanisms that regulate p63 levels. While a unified picture has not emerged, it is clear that the fine-tuning of p63 has evolved to carefully control epithelial cell differentiation and fate.
Collapse
Affiliation(s)
- Kathryn Yoh
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, NY, USA
- *Correspondence: Ron Prywes, Department of Biological Sciences, Columbia University, Fairchild 813A, MC2420, 1212 Amsterdam Avenue, New York, NY 10027, USA,
| |
Collapse
|
58
|
Abstract
It is now emerging that a number of cellular targets of pathogens are involved in the establishment and/or maintenance of epithelial cell polarity. Increasing evidence also suggests that cancer-causing pathogens such as Helicobacter pylori (H. pylori) and human papilloma virus (HPV) may induce oncogenesis by disrupting cell polarity. This is mainly achieved through their ability to deregulate the function of cell polarity components and/or regulators. Hence cell polarity represents the first line of defence against infection. Interestingly, EGFR/RAS oncogenic signals also induce cancer cell invasion by inducing epithelial to mesenchymal transition (EMT). Since the loss of cell polarity is a prerequisition of EMT, cell polarity also represents the last line of defence against cancer cell invasion. As such we argue that cell polarity may be a key defence mechanism against infection and cancer cell invasion. The potential role of cell polarity as a gatekeeper against cancer through its ability to regulate asymmetric cell division and tumour suppression has been discussed in a number of recent reviews. In this review we will focus on the role of cell polarity as a potential target of infection and cancer cell invasion.
Collapse
Affiliation(s)
- Klaus Ebnet
- grid.5949.10000000121729288Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University Münster, Münster, Nordrhein-Westfalen Germany
| | | |
Collapse
|
59
|
Gao K, An J, Zhang Y, Jin X, Ma J, Peng J, Tang Y, Yu L, Zhang P, Wang C. The E3 ubiquitin ligase Itch and Yap1 have antagonistic roles in the regulation of ASPP2 protein stability. FEBS Lett 2014; 589:94-101. [DOI: 10.1016/j.febslet.2014.11.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/28/2014] [Accepted: 11/17/2014] [Indexed: 01/08/2023]
|
60
|
Wang Y, Bu F, Royer C, Serres S, Larkin JR, Soto MS, Sibson NR, Salter V, Fritzsche F, Turnquist C, Koch S, Zak J, Zhong S, Wu G, Liang A, Olofsen PA, Moch H, Hancock DC, Downward J, Goldin RD, Zhao J, Tong X, Guo Y, Lu X. ASPP2 controls epithelial plasticity and inhibits metastasis through β-catenin-dependent regulation of ZEB1. Nat Cell Biol 2014; 16:1092-104. [PMID: 25344754 DOI: 10.1038/ncb3050] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 09/10/2014] [Indexed: 12/16/2022]
Abstract
Epithelial to mesenchymal transition (EMT), and the reverse mesenchymal to epithelial transition (MET), are known examples of epithelial plasticity that are important in kidney development and cancer metastasis. Here we identify ASPP2, a haploinsufficient tumour suppressor, p53 activator and PAR3 binding partner, as a molecular switch of MET and EMT. ASPP2 contributes to MET in mouse kidney in vivo. Mechanistically, ASPP2 induces MET through its PAR3-binding amino-terminus, independently of p53 binding. ASPP2 prevents β-catenin from transactivating ZEB1, directly by forming an ASPP2-β-catenin-E-cadherin ternary complex and indirectly by inhibiting β-catenin's N-terminal phosphorylation to stabilize the β-catenin-E-cadherin complex. ASPP2 limits the pro-invasive property of oncogenic RAS and inhibits tumour metastasis in vivo. Reduced ASPP2 expression results in EMT, and is associated with poor survival in hepatocellular carcinoma and breast cancer patients. Hence, ASPP2 is a key regulator of epithelial plasticity that connects cell polarity to the suppression of WNT signalling, EMT and tumour metastasis.
Collapse
Affiliation(s)
- Yihua Wang
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Fangfang Bu
- 1] International Joint Cancer Institute &Eastern Hospital of Hepatobiliary Surgery, The Second Military Medical University, Shanghai 200433, China [2] PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, 28 Fuxing Road Beijing 100853, China
| | - Christophe Royer
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Sébastien Serres
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7LE, UK
| | - James R Larkin
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7LE, UK
| | - Manuel Sarmiento Soto
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7LE, UK
| | - Nicola R Sibson
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7LE, UK
| | - Victoria Salter
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Florian Fritzsche
- 1] Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK [2] Institute of Surgical Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Casmir Turnquist
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Sofia Koch
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Jaroslav Zak
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Shan Zhong
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Guobin Wu
- Guangxi Cancer Hospital, Guangxi Medical University, Guangxi 530021, China
| | - Anmin Liang
- Guangxi Cancer Hospital, Guangxi Medical University, Guangxi 530021, China
| | - Patricia A Olofsen
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - David C Hancock
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields London WC2A 3LY, UK
| | - Julian Downward
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields London WC2A 3LY, UK
| | - Robert D Goldin
- Centre for Pathology, St Mary's Hospital, Imperial College, London W2 1NY, UK
| | - Jian Zhao
- 1] International Joint Cancer Institute &Eastern Hospital of Hepatobiliary Surgery, The Second Military Medical University, Shanghai 200433, China [2] PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, 28 Fuxing Road Beijing 100853, China
| | - Xin Tong
- 1] International Joint Cancer Institute &Eastern Hospital of Hepatobiliary Surgery, The Second Military Medical University, Shanghai 200433, China [2] PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, 28 Fuxing Road Beijing 100853, China
| | - Yajun Guo
- 1] International Joint Cancer Institute &Eastern Hospital of Hepatobiliary Surgery, The Second Military Medical University, Shanghai 200433, China [2] PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, 28 Fuxing Road Beijing 100853, China
| | - Xin Lu
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
61
|
Nuclear iASPP may facilitate prostate cancer progression. Cell Death Dis 2014; 5:e1492. [PMID: 25341046 PMCID: PMC4649527 DOI: 10.1038/cddis.2014.442] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/11/2014] [Accepted: 08/25/2014] [Indexed: 12/13/2022]
Abstract
One of the major challenges in prostate cancer (PCa) research is the identification of key players that control the progression of primary cancers to invasive and metastatic disease. The majority of metastatic PCa express wild-type p53, whereas loss of p63 expression, a p53 family member, is a common event. Here we identify inhibitor of apoptosis-stimulating protein of p53 (iASPP), a common cellular regulator of p53 and p63, as an important player of PCa progression. Detailed analysis of the prostate epithelium of iASPP transgenic mice, iASPPΔ8/Δ8 mice, revealed that iASPP deficiency resulted in a reduction in the number of p63 expressing basal epithelial cells compared with that seen in wild-type mice. Nuclear and cytoplasmic iASPP expression was greater in PCa samples compared with benign epithelium. Importantly nuclear iASPP associated with p53 accumulation in vitro and in vivo. A pair of isogenic primary and metastatic PCa cell lines revealed that nuclear iASPP is enriched in the highly metastatic PCa cells. Nuclear iASPP is often detected in PCa cells located at the invasive leading edge in vivo. Increased iASPP expression associated with metastatic disease and PCa-specific death in a clinical cohort with long-term follow-up. These results suggest that iASPP function is required to maintain the expression of p63 in normal basal prostate epithelium, and nuclear iASPP may inactivate p53 function and facilitate PCa progression. Thus iASPP expression may act as a predictive marker of PCa progression.
Collapse
|
62
|
Chen Y, Yan W, He S, Chen J, Chen D, Zhang Z, Liu Z, Ding X, Wang A. In vitro effect of iASPP on cell growth of oral tongue squamous cell carcinoma. Chin J Cancer Res 2014; 26:382-90. [PMID: 25232209 DOI: 10.3978/j.issn.1000-9604.2014.07.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 07/24/2014] [Indexed: 01/28/2023] Open
Abstract
iASPP is an inhibitory member of the apoptosis-stimulating proteins of P53 (ASPP) family. iASPP is over expressed in several malignant tumors and potentially affects cancer progression. However, the expression and potential role of iASPP in oral tongue squamous cell carcinoma (OTSCC) have not been addressed. In our study, we detected iASPP expression in OTSCC by immunohistochemistry. iASPP expression is up-regulated in OTSCC tissues. Moreover, in clinical pathology specimens, we found that increased iASPP expression correlates with poor differentiation and lymph node metastasis. Using multicellular tumor spheroids (MTS) and flow cytometry, we demonstrated that iASPP down-regulation arrests OTSCC cells at the G0/G1 phase, induces OTSCC cell apoptosis and inhibits OTSCC cell proliferation. These results indicate that iASPP plays a significant role in the progression of OTSCC and may serve as a biomarker or therapeutic target for OTSCC patients.
Collapse
Affiliation(s)
- Yu Chen
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Wangxiang Yan
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Shuqi He
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Jiechun Chen
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Dan Chen
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Zhaoqiang Zhang
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Zhiguo Liu
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Xueqiang Ding
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| | - Anxun Wang
- 1 Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China, 2 Department of Stomatology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China ; 3 Operation Room, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China ; 4 Department of Stomatology, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China
| |
Collapse
|
63
|
Kim JW, Roh JL, Park Y, Cho KJ, Choi SH, Nam SY, Kim SY. Cytoplasmic iASPP expression as a novel prognostic indicator in oral cavity squamous cell carcinoma. Ann Surg Oncol 2014; 22:662-9. [PMID: 25149434 DOI: 10.1245/s10434-014-4003-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inhibitor of apoptosis-stimulating protein of p53 (iASPP) is a key inhibitor of tumor suppressor p53 that is overexpressed in several human cancers; however, its role in oral cavity squamous cell carcinomas (OSCC) remains unknown. The present study investigated the prognostic role of iASPP in patients with OSCC. METHODS This study included 186 OSCC patients who underwent curative surgery at our institution between 2000 and 2011. Cytoplasmic and nuclear iASPP expression were examined separately by immunohistochemistry, and dichotomized to low and high. Clinicopathological variables associated with locoregional control (LRC), disease-free survival (DFS), and overall survival (OS) were identified by univariate and multivariate analyses. RESULTS Patients were followed-up for a median period of 74 months (range 16-166 months), and 5-year LRC, DFS, and OS was 73.6, 70.2, and 75.3 %, respectively. High iASPP immunostaining reactivity was detected in the cytoplasm and nucleus in 132 (71.0 %) and 93 (50.0 %) patients, respectively. In univariate analysis, pathologic nodal metastasis, advanced overall stage III-IV, lymphovascular invasion, and cytoplasmic iASPP were significantly associated with poor LRC, DFS, and OS (p < 0.05). High-grade and positive resection margins were significant factors associated with poor DFS and OS (p < 0.02). In multivariate analysis, N classification, lymphovascular invasion, and cytoplasmic iASPP expression remained independent variables for LRC, DFS, and OS (p < 0.05). CONCLUSION High iASPP expression in the tumor cell cytoplasm is associated with poor outcomes of OSCC patients in terms of recurrence and survival, suggesting a role for iASPP as a novel biomarker and therapeutic target for OSCC.
Collapse
Affiliation(s)
- Ji Won Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
64
|
STAT1-induced ASPP2 transcription identifies a link between neuroinflammation, cell polarity, and tumor suppression. Proc Natl Acad Sci U S A 2014; 111:9834-9. [PMID: 24958857 DOI: 10.1073/pnas.1407898111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inflammation and loss of cell polarity play pivotal roles in neurodegeneration and cancer. A central question in both diseases is how the loss of cell polarity is sensed by cell death machinery. Here, we identify apoptosis-stimulating protein of p53 with signature sequences of ankyrin repeat-, SH3 domain-, and proline-rich region-containing protein 2 (ASPP2), a haploinsufficient tumor suppressor, activator of p53, and regulator of cell polarity, as a transcriptional target of signal transducer and activator of transcription 1 (STAT1). LPS induces ASPP2 expression in murine macrophage and microglial cell lines, a human monocyte cell line, and primary human astrocytes in vitro. LPS and IFNs induce ASPP2 transcription through an NF-κB RELA/p65-independent but STAT1-dependent pathway. In an LPS-induced maternal inflammation mouse model, LPS induces nuclear ASPP2 in vivo at the blood-cerebral spinal fluid barrier (the brain's barrier to inflammation), and ASPP2 mediates LPS-induced apoptosis. Consistent with the role of ASPP2 as a gatekeeper to inflammation, ASPP2-deficient brains possess enhanced neuroinflammation. Elevated ASPP2 expression is also observed in mouse models and human neuroinflammatory disease tissue, where ASPP2 was detected in GFAP-expressing reactive astrocytes that coexpress STAT1. Because the ability of ASPP2 to maintain cellular polarity is vital to CNS development, our findings suggest that the identified STAT1/ASPP2 pathway may connect tumor suppression and cell polarity to neuroinflammation.
Collapse
|
65
|
Lu M, Miller P, Lu X. Restoring the tumour suppressive function of p53 as a parallel strategy in melanoma therapy. FEBS Lett 2014; 588:2616-21. [PMID: 24844434 DOI: 10.1016/j.febslet.2014.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 01/27/2023]
Abstract
The tumour suppressor p53 is a master sensor of stress and it controls the expression of hundreds to thousands of genes with diverse biological functions including cell cycle arrest, apoptosis, and senescence. Consequently p53 is the most mutated gene found in human cancer and p53 mutation rate varies from 5% to 95%. Importantly p53 activity is often inactivated in tumours expressing structurally wild type p53. Thus one of the major challenges in cancer research is to restore the tumour suppressive function of p53. Intensive studies in the past decade have demonstrated that in addition to mutation, p53 activities are largely regulated by cellular factors that control the expression level and/or transcriptional activities of p53. MDM2, MDM4, p14(ARF) and the ASPP family of proteins are among the most studied regulators of p53. With increased understanding of the complexity of p53 regulation, various p53 reactivating approaches are being developed. This review will focus on the recent understanding of p53 inactivation in melanoma and the approaches to reactivate p53 in preclinical studies. Recent success in the therapeutic targeting of the BRAFV600E oncogenic protein was accompanied with subsequent relapse caused by acquired drug resistance. Restoration of the tumour suppressive function of p53 presents a parallel cancer therapeutic opportunity alongside BRAFV600E inhibition. Thus targeted therapy and concurrent reactivation of p53 may be a fertile ground to achieve synergistic killing of the 50% of cancer cells that express structurally wild type p53.
Collapse
Affiliation(s)
- Min Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Paul Miller
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
66
|
Chikh A, Sanzà P, Raimondi C, Akinduro O, Warnes G, Chiorino G, Byrne C, Harwood CA, Bergamaschi D. iASPP is a novel autophagy inhibitor in keratinocytes. J Cell Sci 2014; 127:3079-93. [PMID: 24777476 DOI: 10.1242/jcs.144816] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The protein iASPP (encoded by PPP1R13L) is an evolutionarily conserved p53 inhibitor, the expression of which is often upregulated in human cancers. We have recently shown that iASPP is a crucial regulator of epidermal homeostasis. Here, we report that iASPP also acts as autophagy inhibitor in keratinocytes. Our data show that depletion of iASPP protects keratinocytes from apoptosis by modulating the expression of Noxa (also known as PMAIP1). In our model, iASPP expression can affect the fission-fusion cycle, mass and shape of mitochondria. iASPP-silenced keratinocytes display disorganization of cytosolic compartments and increased metabolic stress caused by deregulation of mTORC1 signaling. Moreover, increased levels of lipidated LC3 protein confirmed the activation of autophagy in iASPP-depleted cells. We have identified a novel mechanism modulating autophagy in keratinocytes that relies upon iASPP expression specifically reducing the interaction of Atg5-Atg12 with Atg16L1, an interaction that is essential for autophagosome formation or maturation. Using organotypic culture, we further explored the link between autophagy and differentiation, and we showed that impairing autophagy affects epidermal terminal differentiation. Our data provide an alternative mechanism to explain how epithelial integrity is maintained against environmental stressors and might also improve the understanding of the etiology of skin diseases that are characterized by defects in differentiation and DNA damage responses.
Collapse
Affiliation(s)
- Anissa Chikh
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Paolo Sanzà
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Claudio Raimondi
- Centre for Diabetes, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Olufolake Akinduro
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Gary Warnes
- Flow Cytometry Core Facility, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Giovanna Chiorino
- Cancer Genomic Laboratory, Edo ed Elvo Tempia Foundation, 13900 Biella, Italy
| | - Carolyn Byrne
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Catherine A Harwood
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Daniele Bergamaschi
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
67
|
Godin-Heymann N, Wang Y, Slee E, Lu X. Phosphorylation of ASPP2 by RAS/MAPK pathway is critical for its full pro-apoptotic function. PLoS One 2013; 8:e82022. [PMID: 24312625 PMCID: PMC3847091 DOI: 10.1371/journal.pone.0082022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/25/2013] [Indexed: 11/18/2022] Open
Abstract
We reported recently that apoptosis-stimulating protein of p53 (ASPP) 2, an activator of p53, co-operates with oncogenic RAS to enhance the transcription and apoptotic function of p53. However, the detailed mechanism remains unknown. Here we show that ASPP2 is a novel substrate of mitogen-activated protein kinase (MAPK). Phosphorylation of ASPP2 by MAPK is required for RAS-induced increased binding to p53 and increased transactivation of pro-apoptotic genes. In contrast, an ASPP2 phosphorylation mutant exhibits reduced p53 binding and fails to enhance transactivation and apoptosis. Thus phosphorylation of ASPP2 by RAS/MAPK pathway provides a novel link between RAS and p53 in regulating apoptosis.
Collapse
Affiliation(s)
- Nadia Godin-Heymann
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,
| | - Yihua Wang
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,
| | - Elizabeth Slee
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,
| | - Xin Lu
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom,
- * E-mail:
| |
Collapse
|
68
|
Liu X, Li F, Zhao S, Luo Y, Kang J, Zhao H, Yan F, Li S, Ji X. MicroRNA-124-mediated regulation of inhibitory member of apoptosis-stimulating protein of p53 family in experimental stroke. Stroke 2013; 44:1973-80. [PMID: 23696548 DOI: 10.1161/strokeaha.111.000613] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE p53-mediated neuronal death is a central pathway of stroke pathophysiology, but its mechanistic details remain unclear. Here, we identified a novel microRNA mechanism that downregulation of inhibitory member of the apoptosis-stimulating proteins of p53 family (iASPP) by the brain-specific microRNA-124 (miR-124) promotes neuronal death after cerebral ischemia. METHODS In a mouse model of focal permanent cerebral ischemia, the expression of iASPP and miR-124 was quantified by reverse transcription quantitative real-time polymerase chain reaction, immunofluorescence staining, and Western blot. Luciferase reporter assay was used to validate whether miR-124 can directly bind to the 3'-untranslated region of iASPP mRNA. To evaluate the role of miR-124, miR-124 mimic and its inhibitor were transfected into Neuro-2a cells and C57 mice. RESULTS There was no change in the iASPP mRNA level in cerebral ischemia. However, iASPP protein was remarkably decreased, with a concurrent elevation in miR-124 level. Furthermore, miR-124 can bind to the 3'-untranslated region of iASPP in 293T cells and downregulate its protein levels in Neuro-2a cells. In vivo, infusion of miR-124 decreased brain levels of iASPP, whereas inhibition of miR-124 enhanced iASPP levels and significantly reduced infarction in mouse focal cerebral ischemia. CONCLUSIONS These data demonstrate that p53-mediated neuronal cell death after stroke can be nontranscriptionally regulated by a novel mechanism involving suppression of endogenous cell death inhibitors by miR-124. Further dissection of microRNA regulatory mechanisms may lead to new therapeutic opportunities for preventing neuronal death after stroke.
Collapse
Affiliation(s)
- Xiangrong Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Key Laboratory of Neurodegenerative Diseases (Capital Medical University), Ministry of Education, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Janke K, Brockmeier U, Kuhlmann K, Eisenacher M, Nolde J, Meyer HE, Mairbäurl H, Metzen E. Factor inhibiting HIF-1 (FIH-1) modulates protein interactions of apoptosis-stimulating p53 binding protein 2 (ASPP2). J Cell Sci 2013; 126:2629-40. [PMID: 23606740 DOI: 10.1242/jcs.117564] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The asparaginyl hydroxylase factor inhibiting HIF-1 (FIH-1) is an important suppressor of hypoxia-inducible factor (HIF) activity. In addition to HIF-α, FIH-1 was previously shown to hydroxylate other substrates within a highly conserved protein interaction domain, termed the ankyrin repeat domain (ARD). However, to date, the biological role of FIH-1-dependent ARD hydroxylation could not be clarified for any ARD-containing substrate. The apoptosis-stimulating p53-binding protein (ASPP) family members were initially identified as highly conserved regulators of the tumour suppressor p53. In addition, ASPP2 was shown to be important for the regulation of cell polarity through interaction with partitioning defective 3 homolog (Par-3). Using mass spectrometry we identified ASPP2 as a new substrate of FIH-1 but inhibitory ASPP (iASPP) was not hydroxylated. We demonstrated that ASPP2 asparagine 986 (N986) is a single hydroxylation site located within the ARD. ASPP2 protein levels and stability were not affected by depletion or inhibition of FIH-1. However, FIH-1 depletion did lead to impaired binding of Par-3 to ASPP2 while the interaction between ASPP2 and p53, apoptosis and proliferation of the cancer cells were not affected. Depletion of FIH-1 and incubation with the hydroxylase inhibitor dimethyloxalylglycine (DMOG) resulted in relocation of ASPP2 from cell-cell contacts to the cytosol. Our data thus demonstrate that protein interactions of ARD-containing substrates can be modified by FIH-1-dependent hydroxylation. The large cellular pool of ARD-containing proteins suggests that FIH-1 can affect a broad range of cellular functions and signalling pathways under certain conditions, for example, in response to severe hypoxia.
Collapse
Affiliation(s)
- Kirsten Janke
- Institute of Physiology, Faculty of Medicine, University Duisburg-Essen, 45147 Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Molecular mechanisms underlying the interaction of protein phosphatase-1c with ASPP proteins. Biochem J 2013; 449:649-59. [PMID: 23088536 DOI: 10.1042/bj20120506] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The serine/threonine PP-1c (protein phosphatase-1 catalytic subunit) is regulated by association with multiple regulatory subunits. Human ASPPs (apoptosis-stimulating proteins of p53) comprise three family members: ASPP1, ASPP2 and iASPP (inhibitory ASPP), which is uniquely overexpressed in many cancers. While ASPP2 and iASPP are known to bind PP-1c, we now identify novel and distinct molecular interactions that allow all three ASPPs to bind differentially to PP-1c isoforms and p53. iASPP lacks a PP-1c-binding RVXF motif; however, we show it interacts with PP-1c via a RARL sequence with a Kd value of 26 nM. Molecular modelling and mutagenesis of PP-1c-ASPP protein complexes identified two additional modes of interaction. First, two positively charged residues, Lys260 and Arg261 on PP-1c, interact with all ASPP family members. Secondly, the C-terminus of the PP-1c α, β and γ isoforms contain a type-2 SH3 (Src homology 3) poly-proline motif (PxxPxR), which binds directly to the SH3 domains of ASPP1, ASPP2 and iASPP. In PP-1cγ this comprises residues 309-314 (PVTPPR). When the Px(T)PxR motif is deleted or mutated via insertion of a phosphorylation site mimic (T311D), PP-1c fails to bind to all three ASPP proteins. Overall, we provide the first direct evidence for PP-1c binding via its C-terminus to an SH3 protein domain.
Collapse
|
71
|
Kolesinska B, Podwysocka DJ, Rueping MA, Seebach D, Kamena F, Walde P, Sauer M, Windschiegl B, Meyer-Ács M, Vor der Brüggen M, Giehring S. Permeation through Phospholipid Bilayers, Skin-Cell Penetration, Plasma Stability, and CD Spectra ofα- andβ-Oligoproline Derivatives. Chem Biodivers 2013; 10:1-38. [DOI: 10.1002/cbdv.201200393] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Indexed: 11/06/2022]
|
72
|
MicroRNA-124 regulates the proliferation of colorectal cancer cells by targeting iASPP. BIOMED RESEARCH INTERNATIONAL 2013; 2013:867537. [PMID: 23691514 PMCID: PMC3652105 DOI: 10.1155/2013/867537] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/24/2013] [Accepted: 02/27/2013] [Indexed: 01/17/2023]
Abstract
MicroRNAs are a class of small, noncoding RNAs that function as critical regulators of gene expression by targeting mRNAs for translational repression or degradation. In this study, we demonstrate that expression of microRNA-124 (miR-124) is significantly downregulated in colorectal cancer tissues and cell lines, compared to the matched adjacent tissues. We identified and confirmed inhibitor of apoptosis-stimulating protein of p53 (iASPP) as a novel, direct target of miR-124 using target prediction algorithms and luciferase reporter gene assays. Overexpression of miR-124 suppressed iASPP protein expression, upregulated expression of the downstream signaling molecule nuclear factor-kappa B (NF- κ B), and attenuated cell viability, proliferation, and colony formation in SW480 and HT-29 colorectal cancer cells in vitro. Forced overexpression of iASPP partly rescued the inhibitory effect of miR-124 on SW480 and HT29 cell proliferation. Taken together, these findings shed light on the role and mechanism of action of miR-124, indicate that the miR-124/iASPP axis can regulate the proliferation of colorectal cancer cells, and suggest that miR-124 may serve as a potential therapeutic target for colorectal cancer.
Collapse
|
73
|
N terminus of ASPP2 binds to Ras and enhances Ras/Raf/MEK/ERK activation to promote oncogene-induced senescence. Proc Natl Acad Sci U S A 2012; 110:312-7. [PMID: 23248303 DOI: 10.1073/pnas.1201514110] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The ASPP2 (also known as 53BP2L) tumor suppressor is a proapoptotic member of a family of p53 binding proteins that functions in part by enhancing p53-dependent apoptosis via its C-terminal p53-binding domain. Mounting evidence also suggests that ASPP2 harbors important nonapoptotic p53-independent functions. Structural studies identify a small G protein Ras-association domain in the ASPP2 N terminus. Because Ras-induced senescence is a barrier to tumor formation in normal cells, we investigated whether ASPP2 could bind Ras and stimulate the protein kinase Raf/MEK/ERK signaling cascade. We now show that ASPP2 binds to Ras-GTP at the plasma membrane and stimulates Ras-induced signaling and pERK1/2 levels via promoting Ras-GTP loading, B-Raf/C-Raf dimerization, and C-Raf phosphorylation. These functions require the ASPP2 N terminus because BBP (also known as 53BP2S), an alternatively spliced ASPP2 isoform lacking the N terminus, was defective in binding Ras-GTP and stimulating Raf/MEK/ERK signaling. Decreased ASPP2 levels attenuated H-RasV12-induced senescence in normal human fibroblasts and neonatal human epidermal keratinocytes. Together, our results reveal a mechanism for ASPP2 tumor suppressor function via direct interaction with Ras-GTP to stimulate Ras-induced senescence in nontransformed human cells.
Collapse
|
74
|
Wang L, Xing H, Tian Z, Peng L, Li Y, Tang K, Rao Q, Wang M, Wang J. iASPPsv antagonizes apoptosis induced by chemotherapeutic agents in MCF-7 cells and mouse thymocytes. Biochem Biophys Res Commun 2012; 424:414-20. [DOI: 10.1016/j.bbrc.2012.06.124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/25/2012] [Indexed: 12/30/2022]
|
75
|
Elevated expression of iASPP in head and neck squamous cell carcinoma and its clinical significance. Med Oncol 2012; 29:3381-8. [PMID: 22815155 DOI: 10.1007/s12032-012-0306-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 07/04/2012] [Indexed: 01/24/2023]
|
76
|
Patel JB, Patel KD, Patel SR, Shah FD, Shukla SN, Patel PS. Recent Candidate Molecular Markers: Vitamin D Signaling and Apoptosis Specific Regulator of p53 (ASPP) in Breast Cancer. Asian Pac J Cancer Prev 2012; 13:1727-35. [DOI: 10.7314/apjcp.2012.13.5.1727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
77
|
Vangsted A, Klausen TW, Vogel U. Genetic variations in multiple myeloma I: effect on risk of multiple myeloma. Eur J Haematol 2011; 88:8-30. [PMID: 21883480 DOI: 10.1111/j.1600-0609.2011.01700.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Few risk factors have been established for the plasma cell disorder multiple myeloma, but some of these like African American ethnicity and a family history of B-cell lymphoproliferative diseases suggest a genetic component for the disease. Genetic variation represents the genetic basis of variability in a population. The complex interplay between environment and genes for the development of cancer may therefore be influenced by genetic variations. A genetic variation may change the function of the gene, and if the genetic variation is associated with the risk of disease, that particular gene may be involved in the pathogenesis of disease. Genes of interest are genes involved in the normal development and function of the plasma cell and genes that protect us against exposures from the environment, for example, genes involved in the metabolism of xenobiotics, metabolism of folate and methionine, as well as genes involved in inflammation and DNA repair. Identification of genes with potential influence on cancer risk may help us to establish relevant laboratory studies on exposure and dose-response assessment and may help us to test the hypothesis in epidemiological studies. Knowledge of individual at high risk of cancer may offer promising insight for the prevention of cancer.
Collapse
Affiliation(s)
- Annette Vangsted
- Department of Haematology, Roskilde Hospital, Copenhagen University, Roskilde, Denmark.
| | | | | |
Collapse
|
78
|
Differential effects on p53-mediated cell cycle arrest vs. apoptosis by p90. Proc Natl Acad Sci U S A 2011; 108:18937-42. [PMID: 22084066 DOI: 10.1073/pnas.1110988108] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
p53 functions as a central node for organizing whether the cell responds to stress with apoptosis or cell cycle arrest; however, the molecular events that lead to apoptotic responses are not completely understood. Here, we identified p90 (also called Coiled-Coil Domain Containing 8) as a unique regulator for p53. p90 has no obvious effects on either the levels of p53 or p53-mediated cell cycle arrest but is specifically required for p53-mediated apoptosis upon DNA damage. Notably, p90 is crucial for Tip60-dependent p53 acetylation at Lys120, therefore facilitating activation of the proapoptotic targets. These studies indicate that p90 is a critical cofactor for p53-mediated apoptosis through promoting Tip60-mediated p53 acetylation.
Collapse
|
79
|
Wilger DJ, Park JH, Hughes RM, Cuellar ME, Waters ML. Induced-Fit Binding of a Polyproline Helix by a β-Hairpin Peptide. Angew Chem Int Ed Engl 2011; 50:12201-4. [DOI: 10.1002/anie.201106177] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Indexed: 01/23/2023]
|
80
|
Wilger DJ, Park JH, Hughes RM, Cuellar ME, Waters ML. Induced-Fit Binding of a Polyproline Helix by a β-Hairpin Peptide. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201106177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
81
|
Jiang L, Siu MKY, Wong OGW, Tam KF, Lu X, Lam EWF, Ngan HYS, Le XF, Wong ESY, Monteiro LJ, Chan HY, Cheung ANY. iASPP and chemoresistance in ovarian cancers: effects on paclitaxel-mediated mitotic catastrophe. Clin Cancer Res 2011; 17:6924-33. [PMID: 21926165 DOI: 10.1158/1078-0432.ccr-11-0588] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE iASPP is a specific regulator of p53-mediated apoptosis. Herein, we provided the first report on the expression profile of iASPP in ovarian epithelial tumor and its effect on paclitaxel chemosensitivity. EXPERIMENTAL DESIGN Expression and amplification status of iASPP was examined in 203 clinical samples and 17 cell lines using immunohistochemistry, quantitative real-time PCR, and immunoblotting, and correlated with clinicopathologic parameters. Changes in proliferation, mitotic catastrophe, apoptosis, and underlying mechanism in ovarian cancer cells of different p53 status following paclitaxel exposure were also analyzed. RESULTS The protein and mRNA expression of iASPP was found to be significantly increased in ovarian cancer samples and cell lines. High iASPP expression was significantly associated with clear cell carcinoma subtype (P = 0.003), carboplatin and paclitaxel chemoresistance (P = 0.04), shorter overall (P = 0.003), and disease-free (P = 0.001) survival. Multivariate analysis confirmed iASPP expression as an independent prognostic factor. Increased iASPP mRNA expression was significantly correlated with gene amplification (P = 0.023). iASPP overexpression in ovarian cancer cells conferred resistance to paclitaxel by reducing mitotic catastrophe in a p53-independent manner via activation of separase, whereas knockdown of iASPP enhanced paclitaxel-mediated mitotic catastrophe through inactivating separase. Both securin and cyclin B1/CDK1 complex were involved in regulating separase by iASPP. Conversely, overexpressed iASPP inhibited apoptosis in a p53-dependent mode. CONCLUSIONS Our data show an association of iASPP overexpression with gene amplification in ovarian cancer and suggest a role of iASPP in poor patient outcome and chemoresistance, through blocking mitotic catastrophe. iASPP should be explored further as a potential prognostic marker and target for chemotherapy.
Collapse
Affiliation(s)
- Lili Jiang
- Department of Pathology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
iASPP/p63 autoregulatory feedback loop is required for the homeostasis of stratified epithelia. EMBO J 2011; 30:4261-73. [PMID: 21897369 DOI: 10.1038/emboj.2011.302] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 07/22/2011] [Indexed: 11/08/2022] Open
Abstract
iASPP, an inhibitory member of the ASPP (apoptosis stimulating protein of p53) family, is an evolutionarily conserved inhibitor of p53 which is frequently upregulated in human cancers. However, little is known about the role of iASPP under physiological conditions. Here, we report that iASPP is a critical regulator of epithelial development. We demonstrate a novel autoregulatory feedback loop which controls crucial physiological activities by linking iASPP to p63, via two previously unreported microRNAs, miR-574-3p and miR-720. By investigating its function in stratified epithelia, we show that iASPP participates in the p63-mediated epithelial integrity program by regulating the expression of genes essential for cell adhesion. Silencing of iASPP in keratinocytes by RNA interference promotes and accelerates a differentiation pathway, which also affects and slowdown cellular proliferation. Taken together, these data reveal iASPP as a key regulator of epithelial homeostasis.
Collapse
|
83
|
Gillotin S, Lu X. The ASPP proteins complex and cooperate with p300 to modulate the transcriptional activity of p53. FEBS Lett 2011; 585:1778-82. [PMID: 21513714 DOI: 10.1016/j.febslet.2011.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/30/2011] [Accepted: 04/05/2011] [Indexed: 11/25/2022]
Abstract
Understanding how p53 is able to specifically respond to particular stress signals and regulate many different signalling pathways remains a challenge. Several studies have demonstrated that p53's interactions with different protein partners are essential for it to be able to coordinate specific responses. In particular, the apoptotic pathway is regulated by p53 in cooperation with the Apoptosis Stimulating Proteins of p53 (ASPP) proteins. In this study, we showed that the ASPP proteins are able to bind and cooperate with p300, a well defined co-factor of p53, to selectively regulate p53's transcriptional activity on promoters such as p53-inducible gene 3 but not on p21waf1. This is the first demonstration that the ASPPs can function together with p300 in regulating the transcriptional activity of p53.
Collapse
Affiliation(s)
- Sébastien Gillotin
- Ludwig Institute for Cancer Research Ltd., University of Oxford, The Nuffield Department of Clinical Medicine, Oxford, United Kingdom
| | | |
Collapse
|
84
|
Vigneron AM, Ludwig RL, Vousden KH. Cytoplasmic ASPP1 inhibits apoptosis through the control of YAP. Genes Dev 2011; 24:2430-9. [PMID: 21041411 DOI: 10.1101/gad.1954310] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The ASPP (apoptosis-stimulating protein of p53) family of proteins can function in the nucleus to modulate the transcriptional activity of p53, with ASPP1 and ASPP2 contributing to the expression of apoptotic target genes. In this study, we describe a new function for cytoplasmic ASPP1 in controlling YAP (Yes-associated protein)/TAZ. ASPP1 can inhibit the interaction of YAP with LATS1 (large tumor suppressor 1), a kinase that phosphorylates YAP/TAZ and promotes cytoplasmic sequestration and protein degradation. This function of ASPP1 therefore enhances nuclear accumulation of YAP/TAZ and YAP/TAZ-dependent transcriptional regulation. The consequence of YAP/TAZ activation by ASPP1 is to inhibit apoptosis, in part through the down-regulation of Bim expression, leading to resistance to anoikis and enhanced cell migration. These results reveal a potential oncogenic role for cytoplasmic ASPP1, in contrast to the tumor-suppressive activity described previously for nuclear ASPP1.
Collapse
Affiliation(s)
- Arnaud M Vigneron
- The Beatson Institute for Cancer Research, Bearsden, Glasgow G61 1BD, United Kingdom
| | | | | |
Collapse
|
85
|
RNA content in the nucleolus alters p53 acetylation via MYBBP1A. EMBO J 2011; 30:1054-66. [PMID: 21297583 DOI: 10.1038/emboj.2011.23] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 01/11/2011] [Indexed: 12/23/2022] Open
Abstract
A number of external and internal insults disrupt nucleolar structure, and the resulting nucleolar stress stabilizes and activates p53. We show here that nucleolar disruption induces acetylation and accumulation of p53 without phosphorylation. We identified three nucleolar proteins, MYBBP1A, RPL5, and RPL11, involved in p53 acetylation and accumulation. MYBBP1A was tethered to the nucleolus through nucleolar RNA. When rRNA transcription was suppressed by nucleolar stress, MYBBP1A translocated to the nucleoplasm and facilitated p53-p300 interaction to enhance p53 acetylation. We also found that RPL5 and RPL11 were required for rRNA export from the nucleolus. Depletion of RPL5 or RPL11 blocked rRNA export and counteracted reduction of nucleolar RNA levels caused by inhibition of rRNA transcription. As a result, RPL5 or RPL11 depletion inhibited MYBBP1A translocation and p53 activation. Our observations indicated that a dynamic equilibrium between RNA generation and export regulated nucleolar RNA content. Perturbation of this balance by nucleolar stress altered the nucleolar RNA content and modulated p53 activity.
Collapse
|
86
|
RNA interference-mediated silencing of iASPP induces cell proliferation inhibition and G0/G1 cell cycle arrest in U251 human glioblastoma cells. Mol Cell Biochem 2010; 350:193-200. [PMID: 21184255 DOI: 10.1007/s11010-010-0698-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
iASPP is an evolutionally conserved inhibitory member of the ASPP (apoptosis-stimulating protein of p53) protein family. Overexpression of iASPP was observed in several types of human tumors, however, its role in tumorigenesis has not been fully clarified. To investigate the role of iASPP in human glioblastoma multiforme (GMB) progression, the authors employed lentivirus-mediated shRNA to silence endogenous iASPP expression and elucidated iASPP function by analysis of viability, colony formation, DNA synthesis, and cell cycle in p53-mutant glioblastoma cell line U251. iASPP was significantly and sustainably knocked down by iASPP-specific shRNA in U251 cells. Stable down-regulation of iASPP expression-induced cell proliferation inhibition and G0/G1 cell cycle arrest by down-regulation of cyclin D1 and up-regulation of p21(Waf1/Cip1). Thus, the findings not only provide a molecular basis for the role of iASPP in cell cycle progression of glioblastoma cells but also suggest a novel therapeutic target for the treatment of GBM.
Collapse
|
87
|
Alenzi FQ. Cell type specific expression of the apoptosis stimulating protein (ASPP-2) in human tissues. Acta Microbiol Immunol Hung 2010; 57:419-29. [PMID: 21183427 DOI: 10.1556/amicr.57.2010.4.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Apoptosis stimulating proteins of p53 (ASPP-l and ASPP-2) are a novel family of proteins that have been found to co-stimulate p53 activation of Bax (Bcl-2 associated protein X) inducing caspase-mediated apoptosis. Therefore, these proteins may play an important role in regulating apoptosis in normal and neoplastic cells. However, their cellular and tissue distribution has not been documented. The aim of this study was to determine the localization pattern of ASPP-2 in a variety of normal and malignant human tissues, including liver, lung, prostate, small intestine, kidney, ovary, bladder, cervix, breast, stomach, bowel, gallbladder, endometrium, pancreas, spleen and thyroid.The distribution and expression of ASPP-2 was assessed by immunohistochemistry in a range of formalin-fixed, paraffin embedded, benign and malignant human tissues, using a mouse monoclonal antibody against ASPP-2.The results showed a variable pattern of positivity of ASPP-2 within the tissues studied. ASPP-2 expression was localized in the cytoplasmic paranuclear granules in the epithelial cells of most of the organs we studied. The pattern of staining intensity of ASPP-2 correlated to the maturation state in benign tissue and to the differentiation state in the context of bladder cancer.This study indicates that ASPP-2 has a specific distribution pattern within tissues and cells in a way that appears to be related to differentiation. However, the patterns are neither simplistic nor straightforward and will require further investigation in order to appreciate fully their physiological/pathological significance.
Collapse
Affiliation(s)
- Faris Q Alenzi
- Department of Medical Laboratory Sciences, College of Applied Medial Sciences, Al-Kharj University, Al-Kharj, Saudi Arabia.
| |
Collapse
|
88
|
Lu B, Guo H, Zhao J, Wang C, Wu G, Pang M, Tong X, Bu F, Liang A, Hou S, Fan X, Dai J, Wang H, Guo Y. Increased expression of iASPP, regulated by hepatitis B virus X protein-mediated NF-κB activation, in hepatocellular carcinoma. Gastroenterology 2010; 139:2183-2194.e5. [PMID: 20600029 DOI: 10.1053/j.gastro.2010.06.049] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Revised: 05/27/2010] [Accepted: 06/10/2010] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS iASPP is an inhibitory member of the ankyrin-repeat-, SH3-domain- and proline-rich-region-containing protein (ASPP) family; iASPP expression is up-regulated in different human tumor types. We explored the molecular mechanism increased expression of iASPP and its role in hepatocellular carcinoma (HCC). METHODS iASPP expression levels in human liver samples and cell lines were determined by polymerase chain reaction, immunoblot, and immunohistochemical analyses. Luciferase reporter, chromatin immunoprecipitation, and electrophoretic mobility shift assays were used to measure transcriptional activation by nuclear factor-κB (NF-κB). Effects on tumor growth were characterized with MTS, soft agar colony formation, and flow cytometry analyses. Tumorigenicity of cells was studied in nude mice. RESULTS Compared with normal liver cells or tissues, iASPP was expressed at significantly higher levels in HCC cell lines (9/14) and liver samples from patients with HCC, cirrhosis, or hepatitis B virus infection. Increased expression of iASPP was significantly associated with time to recurrence and survival time of patients with HCC. NF-κB activation increased the expression of iASPP through p65/p50 binding to a putative NF-κB-binding site in the iASPP promoter; hepatitis B virus X gene product might up-regulate expression of iASPP. Transgenic expression of iASPP promoted tumor cell proliferation and resistance to chemotherapeutic drugs in vitro and in vivo. CONCLUSIONS iASPP is up-regulated in HCC; it is a direct transcription target of NF-κB. Increased iASPP expression contributes to tumor progression by proliferative and antiapoptotic effects. iASPP might be developed as an HCC therapeutic target or to sensitize cancer cells to chemotherapeutic drugs; it might also be used as a prognostic factor.
Collapse
Affiliation(s)
- Bin Lu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Schlereth K, Charles JP, Bretz AC, Stiewe T. Life or death: p53-induced apoptosis requires DNA binding cooperativity. Cell Cycle 2010; 9:4068-76. [PMID: 20948308 DOI: 10.4161/cc.9.20.13595] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The tumor suppressor p53 provides exquisite protection from cancer by balancing cell survival and death in response to stress. Sustained stress or irreparable damage trigger p53's killer functions to permanently eliminate genetically-altered cells as a potential source of cancer. To prevent the unnecessary loss of cells that could cause premature aging as a result of stem cell attrition, the killer functions of p53 are tightly regulated and balanced against protector functions that promote damage repair and support survival in response to low stress or mild damage. In molecular terms these p53-based cell fate decisions involve protein interactions with cofactors and modifying enzymes, which modulate the activation of distinct sets of p53 target genes. In addition, we demonstrate that part of this regulation occurs at the level of DNA binding. We show that the killer function of p53 requires the four DNA binding domains within the p53 tetramer to interact with one another. These intermolecular interactions enable cooperative binding of p53 to less perfect response elements in the genome, which are present in many target genes essential for apoptosis. Modulating p53 interactions within the tetramer could therefore present a novel promising strategy to fine-tune p53-based cell fate decisions.
Collapse
|
90
|
Cong W, Hirose T, Harita Y, Yamashita A, Mizuno K, Hirano H, Ohno S. ASPP2 regulates epithelial cell polarity through the PAR complex. Curr Biol 2010; 20:1408-14. [PMID: 20619648 DOI: 10.1016/j.cub.2010.06.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 05/21/2010] [Accepted: 06/10/2010] [Indexed: 10/19/2022]
Abstract
The PAR complex, consisting of the evolutionarily conserved PAR-3, PAR-6, and aPKC, regulates cell polarity in many cell types, including epithelial cells [1-4]. Consistently, genetic manipulation of its components affects tissue integrity in multiple biological systems [5-9]. However, the regulatory mechanisms of the PAR complex remain obscure. We report here that apoptosis-stimulating protein of p53 (ASPP2 or TP53BP2), which binds to the tumor suppressor p53 and stimulates its proapoptotic function [10-12], positively regulates epithelial cell polarity by associating with the PAR complex. ASPP2 interacts and colocalizes with PAR-3 at apical cell-cell junctions in the polarized epithelial cells. Depletion of ASPP2 in epithelial cells causes defects in cell polarity, such as the formation of tight junctions and the maintenance and development of apical membrane domains. Moreover, depletion of ASPP2 causes a defect in PAR-3 localization, as well as vice versa. Furthermore, disturbance in the interaction between ASPP2 and PAR-3 causes defects in cell polarity. We conclude that ASPP2 regulates epithelial cell polarity in cooperation with PAR-3 to form an active PAR complex. Our results, taken together with the known functions of ASPP2, suggest a close relationship between cell polarity and other cell regulatory mechanisms mediated by ASPP2.
Collapse
Affiliation(s)
- Weili Cong
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, 3-9 Fuku-ura, Kanazawa-ku, Yokohama, Japan
| | | | | | | | | | | | | |
Collapse
|
91
|
Veksler-Lublinsky I, Shemer-Avni Y, Kedem K, Ziv-Ukelson M. Gene bi-targeting by viral and human miRNAs. BMC Bioinformatics 2010; 11:249. [PMID: 20465802 PMCID: PMC3583137 DOI: 10.1186/1471-2105-11-249] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 05/13/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are an abundant class of small noncoding RNAs (20-24 nts) that can affect gene expression by post-transcriptional regulation of mRNAs. They play important roles in several biological processes (e.g., development and cell cycle regulation). Numerous bioinformatics methods have been developed to identify the function of miRNAs by predicting their target mRNAs. Some viral organisms also encode miRNAs, a fact that contributes to the complex interactions between viruses and their hosts. A need arises to understand the functional relationship between viral and host miRNAs and their effect on viral and host genes. Our approach to meet this challenge is to identify modules where viral and host miRNAs cooperatively regulate host gene expression. RESULTS We present a method to identify groups of viral and host miRNAs that cooperate in post-transcriptional gene regulation, and their target genes that are involved in similar biological processes. We call these groups (genes and miRNAs of human and viral origin) - modules. The modules are found in a new two-stage procedure, which we call bi-targeting, and is presented in this paper. The stages are (i) a new and efficient target prediction, and (ii) a new method for clustering objects of three different data types. In this work we integrate multiple information sources, including miRNA-target binding information, miRNA expression profiles, and GO annotations. Our hypotheses and the methods have been tested on human and Epstein Barr virus (EBV) miRNAs and human genes, for which we found 34 modules. We provide supporting evidence from biological and medical literature for two of our modules. Our code and data are available at http://www.cs.bgu.ac.il/~vaksler/BiTargeting.htm CONCLUSIONS The presented algorithm, which makes use of diverse biological data, is demonstrated to be an efficient approach for finding bi-targeting modules of viral and human miRNAs. These modules can contribute to a better understanding of viral-host interactions and the role that miRNAs play in them.
Collapse
Affiliation(s)
| | - Yonat Shemer-Avni
- Virology and Developmental Genetics/Health Sciences, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Klara Kedem
- Department of Computer Science, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Michal Ziv-Ukelson
- Department of Computer Science, Ben-Gurion University, Beer-Sheva 84105, Israel
| |
Collapse
|
92
|
Hollstein M, Hainaut P. Massively regulated genes: the example of TP53. J Pathol 2010; 220:164-73. [PMID: 19918835 DOI: 10.1002/path.2637] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Intensive study of the TP53 gene over the last three decades has revealed a highly complex network of factors that regulate its performance. The gene has several promoters, alternative splicing occurs and there are alternative translation initiation sites. Up to 10 p53 isoforms have been identified. At the post-translational level, p53 activity depends on its quantity in the cell and on qualitative changes in its structure, intracellular localization, DNA-binding activity and interactions with other proteins. Both accumulation and activation are regulated by an intricate pattern of post-translational modifications, including phosphorylation, acetylation, ubiquitination, sumoylation, neddylation, methylation and glycosylation. The Mdm2 protein, a negative regulator of p53, is the most important determinant of p53 abundance and subcellular localization. Enzymes that post-translationally modify p53 by phosphorylation, methylation and acetylation fine-tune p53 binding to recognition sequences in DNA and p53 interactions with transcription cofactors at promoters of target genes, thereby exerting a discriminatory role in p53 function. This multitude of parameters determining expression, modification, accumulation and localization of p53 proteins may explain how a single gene can display an extensive repertoire of activities. Presumably this is needed, because the p53 protein can have such profound consequences for a cell.
Collapse
|
93
|
Zhao J, Wu G, Bu F, Lu B, Liang A, Cao L, Tong X, Lu X, Wu M, Guo Y. Epigenetic silence of ankyrin-repeat-containing, SH3-domain-containing, and proline-rich-region- containing protein 1 (ASPP1) and ASPP2 genes promotes tumor growth in hepatitis B virus-positive hepatocellular carcinoma. Hepatology 2010; 51:142-53. [PMID: 20034025 DOI: 10.1002/hep.23247] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UNLABELLED The ankyrin-repeat-containing, SH3-domain-containing, and proline-rich-region-containing protein (ASPP) family of proteins regulates apoptosis through interaction with p53 and its family members. This study evaluated the epigenetic regulation of ASPP1 and ASPP2 in hepatitis B virus (HBV)-positive hepatocellular carcinoma (HCC) and explores the effects of down-regulation of ASPP1 and ASPP2 on the development of HCC. HCC cell lines and tissues from HCC patients were used to examine the expression and methylation of ASPP1 and ASPP2. The expression of ASPP1 and ASPP2 was diminished in HCC cells by epigenetic silence owing to hypermethylation of ASPP1 and ASPP2 promoters. Analyses of 51 paired HCC and surrounding nontumor tissues revealed that methylation of ASPP1 and ASPP2 was associated with the decreased expression of ASPP1 and ASPP2 in tumor tissues and the early development of HCC. Moreover, ASPP2 became methylated upon HBV x protein (HBx) expression. The suppressive effects on tumor growth by ASPP1 and ASPP2 were examined with RNA interference-mediated gene silence. Down-regulation of ASPP1 and ASPP2 promoted the growth of HCC cells in soft agar and in nude mice and decreased the sensitivity of HCC cells to apoptotic stimuli. CONCLUSION ASPP1 and ASPP2 genes are frequently down-regulated by DNA methylation in HBV-positive HCC, which may play important roles in the development of HCC. These findings provide new insight into the molecular mechanisms leading to hepatocarcinogenesis and may have potent therapeutic applications.
Collapse
Affiliation(s)
- Jian Zhao
- International Joint Cancer Institute & Eastern Hospital of Hepatobiliary Surgery, Second Military Medical University, Shanghai, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Laska MJ, Lowe SW, Zender L, Hearn S, Vogel U, Jensen UB, Bric A, Nexø BA. Enforced expression of PPP1R13L increases tumorigenesis and invasion through p53-dependent and p53-independent mechanisms. Mol Carcinog 2009; 48:832-42. [PMID: 19263435 PMCID: PMC3328301 DOI: 10.1002/mc.20528] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PPP1R13L was initially identified as a protein that binds to the NF-kappaB subunit p65/RelA and inhibits its transcriptional activity. It also binds p53 and inhibits its action. One set of experimental findings based on overexpression of PPP1R13L indicates that PPP1R13L blocks apoptosis. Another set of experiments, based on endogenous production of PPP1R13L, suggests that the protein may sometimes be pro-apoptotic. We have used primary mouse embryonic fibroblasts (MEFs), dually transformed by HRAS and adenovirus E1A and differing in their p53 status, to explore the effects of PPP1R13L overexpression, thus examining the ability of PPP1R13L to act as an oncoprotein. We found that overexpression of PPP1R13L strongly accelerated tumor formation by RAS/E1A. PPP1R13L overexpressing cells were depleted for both p53 and active p65/RelA and we found that both p53-dependent and -independent apoptosis pathways were modulated by PPP1R13L. Finally, studies with the proteasome inhibitor MG132 revealed that overexpression of PPP1R13L causes faster p53 degradation, a likely explanation for the depletion of p53. Taken together, our results show that increased levels of PPP1R13L can increase tumorigenesis and furthermore suggest that PPP1R13L can influence metastasis.
Collapse
|
95
|
Gillotin S. iASPP, A potential drug target in cancer therapy. Leuk Res 2009; 33:1175-7. [DOI: 10.1016/j.leukres.2009.04.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 04/26/2009] [Accepted: 04/28/2009] [Indexed: 10/20/2022]
|
96
|
Construction of a full-length iASPP expression plasmid pcDNA3.1(+)/iASPP and its biological activity. Plasmid 2009; 62:10-15. [DOI: 10.1016/j.plasmid.2009.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
97
|
Liu ZJ, Cai Y, Hou L, Gao X, Xin HM, Lu X, Zhong S, Gu SZ, Chen J. Effect of RNA Interference of iASPP on the Apoptosis in MCF-7 Breast Cancer Cells. Cancer Invest 2009; 26:878-82. [DOI: 10.1080/07357900801965042] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
98
|
Abstract
The traditional view of p53 activation includes three steps-p53 stabilization, DNA binding, and transcriptional activation. However, recent studies indicate that each step of p53 activation is more complex than originally anticipated. Moreover, both genetic studies in mice and in vitro studies with purified components suggest that the classical model may not be sufficient to explain all aspects of p53 activation in vivo. To reconcile these differences, we propose that antirepression, the release of p53 from repression by factors such as Mdm2 and MdmX, is a key step in the physiological activation of p53.
Collapse
Affiliation(s)
- Jan-Philipp Kruse
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | | |
Collapse
|
99
|
Uhlmann-Schiffler H, Kiermayer S, Stahl H. The DEAD box protein Ddx42p modulates the function of ASPP2, a stimulator of apoptosis. Oncogene 2009; 28:2065-73. [PMID: 19377511 DOI: 10.1038/onc.2009.75] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ddx42p is a recently characterized mammalian DEAD box protein with unknown cellular function. We found that in human cells Ddx42p physically interacts with ASPP2, a major apoptosis inducer known to enhance p53 transactivation of proapoptotic genes. The proteins interact via a domain within the carboxy-terminal part of Ddx42p and a mid-amino-terminal sequence as well as the ankyrin-SH3 region of ASPP2. Overexpression of Ddx42p interferes with apoptosis induction by ASPP2, whereas Ddx42p knockdown reduces the survival rate of cultured human cells. In addition, ASPP2 is found in cytoplasm and nucleus at low Ddx42p level, and predominantly in cytoplasm at high concentration of Ddx42p, respectively. Our results show that Ddx42p is capable of modulating ASPP2 function.
Collapse
Affiliation(s)
- H Uhlmann-Schiffler
- Department of Medical Biochemistry and Molecular Biology, The Saarland University, Homburg, Germany.
| | | | | |
Collapse
|
100
|
Tian C, Xing G, Xie P, Lu K, Nie J, Wang J, Li L, Gao M, Zhang L, He F. KRAB-type zinc-finger protein Apak specifically regulates p53-dependent apoptosis. Nat Cell Biol 2009; 11:580-91. [PMID: 19377469 DOI: 10.1038/ncb1864] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 02/09/2009] [Indexed: 12/27/2022]
Abstract
Only a few p53 regulators have been shown to participate in the selective control of p53-mediated cell cycle arrest or apoptosis. How p53-mediated apoptosis is negatively regulated remains largely unclear. Here we report that Apak (ATM and p53-associated KZNF protein), a Krüppel-associated box (KRAB)-type zinc-finger protein, binds directly to p53 in unstressed cells, specifically downregulates pro-apoptotic genes, and suppresses p53-mediated apoptosis by recruiting KRAB-box-associated protein (KAP)-1 and histone deacetylase 1 (HDAC1) to attenuate the acetylation of p53. Apak inhibits p53 activity by interacting with ATM, a previously identified p53 activator. In response to stress, Apak is phosphorylated by ATM and dissociates from p53, resulting in activation of p53 and induction of apoptosis. These findings revealed Apak to be a negative regulator of p53-mediated apoptosis and showed the dual role of ATM in p53 regulation.
Collapse
Affiliation(s)
- Chunyan Tian
- State Key Laboratory of Proteomics, Beijing Proteomics Research Center, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|