51
|
Mattison SA, Blatch GL, Edkins AL. HOP expression is regulated by p53 and RAS and characteristic of a cancer gene signature. Cell Stress Chaperones 2017; 22:213-223. [PMID: 27987076 PMCID: PMC5352595 DOI: 10.1007/s12192-016-0755-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/21/2016] [Accepted: 12/07/2016] [Indexed: 01/01/2023] Open
Abstract
The Hsp70/Hsp90 organising protein (HOP) is a co-chaperone essential for client protein transfer from Hsp70 to Hsp90 within the Hsp90 chaperone machine. Although HOP is upregulated in various cancers, there is limited information from in vitro studies on how HOP expression is regulated in cancer. The main objective of this study was to identify the HOP promoter and investigate its activity in cancerous cells. Bioinformatic analysis of the -2500 to +16 bp region of the HOP gene identified a large CpG island and a range of putative cis-elements. Many of the cis-elements were potentially bound by transcription factors which are activated by oncogenic pathways. Luciferase reporter assays demonstrated that the upstream region of the HOP gene contains an active promoter in vitro. Truncation of this region suggested that the core HOP promoter region was -855 to +16 bp. HOP promoter activity was highest in Hs578T, HEK293T and SV40- transformed MEF1 cell lines which expressed mutant or inactive p53. In a mutant p53 background, expression of wild-type p53 led to a reduction in promoter activity, while inhibition of wild-type p53 in HeLa cells increased HOP promoter activity. Additionally, in Hs578T and HEK293T cell lines containing inactive p53, expression of HRAS increased HOP promoter activity. However, HRAS activation of the HOP promoter was inhibited by p53 overexpression. These findings suggest for the first time that HOP expression in cancer may be regulated by both RAS activation and p53 inhibition. Taken together, these data suggest that HOP may be part of the cancer gene signature induced by a combination of mutant p53 and mutated RAS that is associated with cellular transformation.
Collapse
Affiliation(s)
- Stacey A Mattison
- Biomedical Biotechnology Research Unit, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - Gregory L Blatch
- Biomedical Biotechnology Research Unit, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Adrienne L Edkins
- Biomedical Biotechnology Research Unit, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa.
| |
Collapse
|
52
|
Chen YY, Chan KM. Differential effects of metal ions on TCDD-induced cytotoxicity and cytochrome P4501A1 gene expression in a zebrafish liver (ZFL) cell-line. Metallomics 2016; 8:236-51. [PMID: 26612010 DOI: 10.1039/c5mt00219b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Trace metal ions and trace organic compounds are common co-contaminants in the environment that pose risks to human health. We evaluated the effects of four metal ions (As(3+), Cu(2+), Hg(2+), and Zn(2+)) on 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced cytotoxicity and the expression of the cytochrome P4501A1 gene (cyp1a1) in the zebrafish liver (ZFL) cell line. A metal accumulation study showed that Cu and Zn did not accumulate in ZFL cells. However, As and Hg did accumulate, which resulted in the inhibition of TCDD-mediated induction of cyp1a1 mRNA and protein expression, and 7-ethoxyresorufin O-deethylase activity. A luciferase assay showed that both As(3+) and Hg(2+) inhibited the TCDD-induced activity of gene constructs containing either synthetic 3XRE or a distal cyp1a1 promoter region, implying that the decreased levels of TCDD-induced cyp1a1 were due to transcriptional effects. A proteomic study showed that the toxic effects of As(3+) might be due to changes in cellular metabolic processes, the cellular stimulation response and the cellular redox state in ZFL cells.
Collapse
Affiliation(s)
- Ying Ying Chen
- School of Life Sciences, Chinese University, Sha Tin, Hong Kong SAR, China.
| | - King Ming Chan
- School of Life Sciences, Chinese University, Sha Tin, Hong Kong SAR, China.
| |
Collapse
|
53
|
Huang Y, Li H, Wang L, Mao X, Li G. Highly Sensitive Protein Detection Based on Smart Hybrid Nanocomposite-Controlled Switch of DNA Polymerase Activity. ACS APPLIED MATERIALS & INTERFACES 2016; 8:28202-28207. [PMID: 27681499 DOI: 10.1021/acsami.6b09270] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
In this work, we have successfully designed a smart and flexible signal amplification method based on a newly synthesized hybrid nanocomposite with switchable enzyme activity for specific and sensitive protein detection. The smart hybrid nanocomposite synthesized here is initially loaded with quenched fluorophore and a unique aptamer-inhibited DNA polymerase. It then undergoes target protein-triggered release of the fluorophore and activation of the DNA polymerase, which can thereby promote multiple catalytic reactions and recycled use of the target protein, resulting in the generation of highly amplified signals. Therefore, a small amount of target protein can lead to a large amount of signal without being consumed. In addition, the programmable control of DNA polymerase activity may effectively reduce background signal and avoid false positive results, which may further facilitate an efficient detection of small amounts of protein. By taking the detection of human stress-induced phosphoprotein 1 (STIP1) as an example, the excellent performance of this method has been verified. Furthermore, the proposed method has been used to analyze serum STIP1 from patients of ovarian cancer, showing promising application in clinical practice.
Collapse
Affiliation(s)
- Yue Huang
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University , Nanjing 210093, P.R. China
| | - Hao Li
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University , Nanjing 210093, P.R. China
| | - Lei Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University , Nanjing 210093, P.R. China
| | - Xiaoxia Mao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University , Shanghai 200444, P.R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biochemistry, Nanjing University , Nanjing 210093, P.R. China
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University , Shanghai 200444, P.R. China
| |
Collapse
|
54
|
Wang W, Liu Y, Zhao Z, Xie C, Xu Y, Hu Y, Quan H, Lou L. Y-632 inhibits heat shock protein 90 (Hsp90) function by disrupting the interaction between Hsp90 and Hsp70/Hsp90 organizing protein, and exerts antitumor activity in vitro and in vivo. Cancer Sci 2016; 107:782-90. [PMID: 27002306 PMCID: PMC4968598 DOI: 10.1111/cas.12934] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 12/16/2022] Open
Abstract
Heat shock protein 90 (Hsp90) stabilizes a variety of proteins required for cancer cell survival and has been identified as a promising drug target for cancer treatment. To date, several Hsp90 inhibitors have entered into clinical trials, but none has been approved for cancer therapy yet. Thus, exploring new Hsp90 inhibitors with novel mechanisms of action is urgent. In the present study, we show that Y-632, a novel pyrimidine derivative, inhibited Hsp90 in a different way from the conventional Hsp90 inhibitor geldanamycin. Y-632 induced degradation of diverse Hsp90 client proteins through the ubiquitin-proteasome pathway, as geldanamycin did; however, it neither directly bound to Hsp90 nor inhibited Hsp90 ATPase activity. Y-632 inhibited Hsp90 function mainly through inducing intracellular thiol oxidation, which led to disruption of the Hsp90-Hsp70/Hsp90 organizing protein complex and further induced cell adhesion inhibition, G0 /G1 cell cycle arrest, and apoptosis. Moreover, Y-632 efficiently overcame imatinib resistance mediated by Bcr-Abl point mutations both in vitro and in vivo. We believe that Y-632, acting as a novel small-molecule inhibitor of the Hsp90-Hsp70/Hsp90 organizing protein complex, has great potential to be a promising Hsp90 inhibitor for cancer therapy, such as for imatinib-resistant leukemia.
Collapse
Affiliation(s)
- Wenqian Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yang Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhixin Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chengying Xie
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yongping Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Youhong Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haitian Quan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Liguang Lou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
55
|
de Lacerda TCS, Costa-Silva B, Giudice FS, Dias MVS, de Oliveira GP, Teixeira BL, Dos Santos TG, Martins VR. Prion protein binding to HOP modulates the migration and invasion of colorectal cancer cells. Clin Exp Metastasis 2016; 33:441-51. [PMID: 27112151 DOI: 10.1007/s10585-016-9788-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed malignancies. The generation of conventional treatments has improved, but approximately 50 % of patients with CRC who undergo potentially curative surgery ultimately relapse and die, usually as a consequence of metastatic disease. Our previous findings showed that engagement of the cellular prion protein (PrP(C)) to its ligand HSP70/90 heat shock organizing protein (HOP) induces proliferation of glioblastomas. In addition, PrP(C) has been described as an important modulator of colorectal tumor growth. Here, we investigated the biological relevance of the PrP(C)-HOP interaction in CRC cells. We demonstrate that HOP induced the migration and invasion of CRC cell lines in a PrP(C)-dependent manner and that phosphorylation of the ERK1/2 pathway is a downstream mediator of these effects. Additionally, we show that a HOP peptide with the ability to bind PrP(C) and abolish the PrP(C)-HOP interaction inhibited the migration and invasion of CRC cells. Together, these data indicate that the disruption of the PrP(C)-HOP complex could be a potential therapeutic target for modulating the migratory and invasive cellular properties that lead to metastatic CRC.
Collapse
Affiliation(s)
- Tonielli Cristina Sousa de Lacerda
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.,Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Av. Prof. Lineu Prestes, 748 - Cidade Universitária, São Paulo, SP, 05508-000, Brazil
| | - Bruno Costa-Silva
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.,Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, 10021, USA.,Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasilia, Doca de Pedrouços, 1400-038, Lisbon, Portugal
| | - Fernanda Salgueiredo Giudice
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Marcos Vinicios Salles Dias
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Gabriela Pintar de Oliveira
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Bianca Luise Teixeira
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Tiago Goss Dos Santos
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Vilma Regina Martins
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.
| |
Collapse
|
56
|
Immunoproteasome deficiency is a feature of non-small cell lung cancer with a mesenchymal phenotype and is associated with a poor outcome. Proc Natl Acad Sci U S A 2016; 113:E1555-64. [PMID: 26929325 DOI: 10.1073/pnas.1521812113] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The immunoproteasome plays a key role in generation of HLA peptides for T cell-mediated immunity. Integrative genomic and proteomic analysis of non-small cell lung carcinoma (NSCLC) cell lines revealed significantly reduced expression of immunoproteasome components and their regulators associated with epithelial to mesenchymal transition. Low expression of immunoproteasome subunits in early stage NSCLC patients was associated with recurrence and metastasis. Depleted repertoire of HLA class I-bound peptides in mesenchymal cells deficient in immunoproteasome components was restored with either IFNγ or 5-aza-2'-deoxycytidine (5-aza-dC) treatment. Our findings point to a mechanism of immune evasion of cells with a mesenchymal phenotype and suggest a strategy to overcome immune evasion through induction of the immunoproteasome to increase the cellular repertoire of HLA class I-bound peptides.
Collapse
|
57
|
Zhang W, Niu C, He W, Hou T, Sun X, Xu L, Zhang Y. Upregulation of centrosomal protein 55 is associated with unfavorable prognosis and tumor invasion in epithelial ovarian carcinoma. Tumour Biol 2015; 37:6239-54. [PMID: 26615423 PMCID: PMC4875171 DOI: 10.1007/s13277-015-4419-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022] Open
Abstract
Centrosomal protein 55 (CEP55) is a cell cycle regulator implicated in development of certain cancers. However, characteristics of CEP55 expression and its clinical/prognostic significance are unclear in human epithelial ovarian carcinoma (EOC). Therefore, we investigated the expression and clinicopathological significance of CEP55 in patients with EOC and its role in regulating invasion and metastasis of ovarian cell lines. CEP55 mRNA and protein expression levels were detected by quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry (IHC). Potential associations of CEP55 expression scores with clinical parameters and patient survival were evaluated. CEP55 function was investigated further using RNA interference, wound healing assay, transwell assay, immunofluorescence analysis, qRT-PCR, and Western blotting. CEP55 was significantly upregulated in ovarian cancer cell lines and lesions compared with normal cells and adjacent noncancerous ovarian tissues. In the 213 EOC samples, CEP55 protein levels were positively correlated with clinical stage (P < 0.001), lymph node metastasis (P < 0.001), intraperitoneal metastasis (P < 0.001), tumor recurrence (P < 0.001), differentiation grade (P < 0.001), residual tumor size (P < 0.001), ascites see tumor cells (P = 0.020), and serum CA153 level (P < 0.001). Moreover, patients with aberrant CEP55 protein expression showed tendencies to receive neoadjuvant chemotherapy (P < 0.001) and cytoreductive surgery (P = 0.020). By contrast, no significant correlation was detected between the protein levels and patient age, histological type, or serum CA125, CA199, CA724, NSE, CEA, and β-HCG levels. Patients with high CEP55 protein expression had shorter overall survival and disease-free survival compared with those with low CEP55 expression. Multivariate analysis implicated CEP55 as an independent prognostic indicator for EOC patients. Additionally, downregulation of CEP55 in ovarian cancer cells remarkably inhibited cellular motility and invasion. Aberrant CEP55 expression may predict unfavorable clinical outcomes in EOC patients and play an important role in regulating invasion in ovarian cancer cells. Thus, CEP55 may serve as a prognostic marker and therapeutic target for EOC.
Collapse
Affiliation(s)
- Weijing Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecologic Oncology, Cancer Center, Sun Yat-Sen University, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Chunhao Niu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecologic Oncology, Cancer Center, Sun Yat-Sen University, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Weiling He
- Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080, People's Republic of China
| | - Teng Hou
- Department of Urology, Wuhan Union Hospital of Huazhong University of Science and Technology, No.1277, Han Kou Jie Fang Road, Wuhan, 430022, People's Republic of China
| | - Xiaoying Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecologic Oncology, Cancer Center, Sun Yat-Sen University, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Liqun Xu
- Department of Gynecology, Women and Children Hospital of Guangdong Province, No.13, Guang Yuan Road, Guangzhou, 510060, People's Republic of China
| | - Yanna Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecologic Oncology, Cancer Center, Sun Yat-Sen University, No. 651, Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
58
|
Wang Y, Shan Q, Hou G, Zhang J, Bai J, Lv X, Xie Y, Zhu H, Su S, Li Y, Zi J, Lin L, Han W, Zhao X, Wang H, Xu N, Wu L, Lou X, Liu S. Discovery of potential colorectal cancer serum biomarkers through quantitative proteomics on the colonic tissue interstitial fluids from the AOM-DSS mouse model. J Proteomics 2015; 132:31-40. [PMID: 26581642 DOI: 10.1016/j.jprot.2015.11.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/25/2015] [Accepted: 11/10/2015] [Indexed: 01/29/2023]
Abstract
UNLABELLED Quantitative proteomic analysis was performed using iTRAQ to discover colorectal cancer (CRC)-related proteins in tissue interstitial fluids (TIFs). A typical inflammation-related CRC mouse model was generated using azoxymethane-dextran sodium sulfate (AOM-DSS), and TIFs were collected from these mice in four stages during CRC development. Using stringent criteria, a total of 144 proteins displayed changes in their abundances during tumor growth, including 45 that consecutively increased, 17 that consecutively decreased and 82 that changed irregularly. Of these 144 proteins, 24 of the consecutively changed proteins were measured using MRM in individual TIF samples, and 18 were verified. Twelve proteins verified to be consecutively increased in TIFs were examined using MRM to evaluate changes in their abundance in individual mouse serum samples. The abundances of leucine-rich alpha-2-glycoprotein 1 (LRG1), tubulin beta-5 chain (TUBB5) and immunoglobulin J chain (IGJ) were significantly higher in CRC mice than in control mice. Using clinical samples and MRM, we further verified that LRG1 and TUBB5 are potential CRC serum biomarkers. These data demonstrate that coupling dynamic TIF proteomics with targeted serum proteomics in an animal model is a promising avenue for pursuing the discovery of tumor serum biomarkers. BIOLOGICAL SIGNIFICANCE Colorectal cancer (CRC) is one of the most dangerous diseases worldwide. However, few of CRC biomarkers possess satisfied specificity and sensitivity in clinical practices. Exploration of more CRC biomarkers, especially in serum, is an urgent and also a time-consuming campaign in the CRC study. Our study demonstrates that quantitatively evaluating the phase-dependent proteins in colonic tissue interstitial fluids from AOM-DSS mice is a feasible and effective way for exploration of the CRC-related proteins and the potential serum biomarkers. We identified two proteins, LRG1 and TUBB5, which may be practicable in human clinical samples as CRC serum biomarkers. To sum up, this study provides a novel angle to explore the critical factors in tumorigenesis and a new pipeline for potential serum biomarker discovery and verification.
Collapse
Affiliation(s)
- Yang Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiang Shan
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Guixue Hou
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ju Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaolei Lv
- Beijing Protein Innovation, Beijing 101318, China
| | - Yingying Xie
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huishan Zhu
- Beijing Protein Innovation, Beijing 101318, China
| | - Siyuan Su
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Li
- Beijing Protein Innovation, Beijing 101318, China
| | - Jin Zi
- Proteomics Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Liang Lin
- Proteomics Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Wenxiao Han
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Lin Wu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Xiaomin Lou
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Siqi Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Proteomics Division, BGI-Shenzhen, Shenzhen, Guangdong 518083, China.
| |
Collapse
|
59
|
Beraldo FH, Thomas A, Kolisnyk B, Hirata PH, De Jaeger X, Martyn AC, Fan J, Goncalves DF, Cowan MF, Masood T, Martins VR, Gros R, Prado VF, Prado MAM. Hyperactivity and attention deficits in mice with decreased levels of stress-inducible phosphoprotein 1 (STIP1). Dis Model Mech 2015; 8:1457-66. [PMID: 26398952 PMCID: PMC4631792 DOI: 10.1242/dmm.022525] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/04/2015] [Indexed: 12/21/2022] Open
Abstract
Stress-inducible phosphoprotein I (STIP1, STI1 or HOP) is a co-chaperone intermediating Hsp70/Hsp90 exchange of client proteins, but it can also be secreted to trigger prion protein-mediated neuronal signaling. Some mothers of children with autism spectrum disorders (ASD) present antibodies against certain brain proteins, including antibodies against STIP1. Maternal antibodies can cross the fetus blood-brain barrier during pregnancy, suggesting the possibility that they can interfere with STIP1 levels and, presumably, functions. However, it is currently unknown whether abnormal levels of STIP1 have any impact in ASD-related behavior. Here, we used mice with reduced (50%) or increased STIP1 levels (fivefold) to test for potential ASD-like phenotypes. We found that increased STIP1 regulates the abundance of Hsp70 and Hsp90, whereas reduced STIP1 does not affect Hsp70, Hsp90 or the prion protein. Interestingly, BAC transgenic mice presenting fivefold more STIP1 show no major phenotype when examined in a series of behavioral tasks, including locomotor activity, elevated plus maze, Morris water maze and five-choice serial reaction time task (5-CSRTT). In contrast, mice with reduced STIP1 levels are hyperactive and have attentional deficits on the 5-CSRTT, but exhibit normal performance for the other tasks. We conclude that reduced STIP1 levels can contribute to phenotypes related to ASD. However, future experiments are needed to define whether it is decreased chaperone capacity or impaired prion protein signaling that contributes to these phenotypes. Summary: Here, using a series of behavioral tests including touchscreen tasks we show that decreased levels of stress-inducible phosphoprotein 1 (STIP1) lead to attention deficits and hyperactivity in mice.
Collapse
Affiliation(s)
- Flavio H Beraldo
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Anu Thomas
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Benjamin Kolisnyk
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada Program in Neuroscience, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Pedro H Hirata
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Xavier De Jaeger
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Amanda C Martyn
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Jue Fan
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Daniela F Goncalves
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Matthew F Cowan
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Talal Masood
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada Program in Neuroscience, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Vilma R Martins
- Department of Molecular and Cell Biology, International Research Center, A.C. Camargo Cancer Center and National Institute for Translational Neuroscience Research Center, Sao Paulo, SP 01508-010, Brazil
| | - Robert Gros
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Vania F Prado
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada Program in Neuroscience, The University of Western Ontario, London, Ontario N6A5B7, Canada Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A5B7, Canada Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A5B7, Canada
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A5B7, Canada Program in Neuroscience, The University of Western Ontario, London, Ontario N6A5B7, Canada Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A5B7, Canada Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A5B7, Canada
| |
Collapse
|
60
|
Chao A, Tsai CL, Jung SM, Chuang WC, Kao C, Hsu A, Chen SH, Lin CY, Lee YC, Lee YS, Wang TH, Wang HS, Lai CH. BAI1-Associated Protein 2-Like 1 (BAIAP2L1) Is a Potential Biomarker in Ovarian Cancer. PLoS One 2015. [PMID: 26222696 PMCID: PMC4519316 DOI: 10.1371/journal.pone.0133081] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Brain-specific angiogenesis inhibitor 1 (BAI1)-associated protein 2-like 1 (BAIAP2L1), also known as insulin receptor tyrosine kinase substrate (IRTKS), is involved in plasma membrane protrusion and actin formation during cell morphogenesis and migration. BAIAP2L1 is recently reported to promote cell proliferation through activation of the EGFR-ERK pathway in hepatocellular carcinoma. In this study, we report the first comprehensive study of BAIAP2L1 upregulation in human ovarian cancer. Upregulation of BAIAP2L1 in ovarian tumors was first found during RNA screening and confirmed by immunohistochemical studies on ovarian cancers and other cancer types. Significant upregulation of BAIAP2L1 in ovarian cancer was validated by analyzing multiple independent cohorts in publicly available data sets. Furthermore, BAIAP2L1 protein expression in metastatic lesions was higher than the corresponding primary tumors. Functional assays in ovarian cancer cells revealed that BAIAP2L1 is involved in promoting cell proliferation and avoiding apoptosis. In conclusion, results of this study not only indicate that BAIAP2L1 can be used as a biomarker for human ovarian cancer but also reveal its role in cancer biology. Further elucidation of the role of BAIAP2L1 in context of the insulin receptor signaling pathways of cancer cells is warranted for developing cancer therapeutics by targeting cancer-specific metabolism.
Collapse
Affiliation(s)
- Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chia-Lung Tsai
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Ming Jung
- Department of Clinical Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Wei-Chi Chuang
- Graduate Institute of Biomedical Science, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chieh Kao
- Graduate Institute of Biomedical Science, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - An Hsu
- Graduate Institute of Biomedical Science, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shun-Hua Chen
- Graduate Institute of Biomedical Science, School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chao Lee
- College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yun-Shien Lee
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
- * E-mail: (THW); (YSL)
| | - Tzu-Hao Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (THW); (YSL)
| | - Hsin-Shih Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chyong-Huey Lai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
61
|
Wagner M, Wiig H. Tumor Interstitial Fluid Formation, Characterization, and Clinical Implications. Front Oncol 2015; 5:115. [PMID: 26075182 PMCID: PMC4443729 DOI: 10.3389/fonc.2015.00115] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/06/2015] [Indexed: 12/18/2022] Open
Abstract
The interstitium, situated between the blood and lymph vessels and the cells, consists of a solid or matrix phase and a fluid phase representing the tissue microenvironment. In the present review, we focus on the interstitial fluid phase of solid tumors, the tumor interstitial fluid (TIF), i.e., the fluid bathing the tumor and stroma cells, also including immune cells. This is a component of the internal milieu of a solid tumor that has attracted regained attention. Access to this space may provide important insight into tumor development and therapy response. TIF is formed by transcapillary filtration, and since this fluid is not readily available we discuss available techniques for TIF isolation, results from subsequent characterization and implications of recent findings with respect to fluid filtration and uptake of macromolecular therapeutic agents. There appear to be local gradients in signaling substances from neoplastic tissue to plasma that may provide new understanding of tumor biology. The development of sensitive proteomic technologies has made TIF a valuable source for tumor specific proteins and biomarker candidates. Potential biomarkers will appear locally in high concentrations in tumors and may eventually be found diluted in the plasma. Access to TIF that reliably reflects the local tumor microenvironment enables identification of substances that can be used in early detection and monitoring of disease.
Collapse
Affiliation(s)
- Marek Wagner
- Department of Biomedicine, University of Bergen , Bergen , Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen , Bergen , Norway
| |
Collapse
|
62
|
Baindur-Hudson S, Edkins AL, Blatch GL. Hsp70/Hsp90 organising protein (hop): beyond interactions with chaperones and prion proteins. Subcell Biochem 2015; 78:69-90. [PMID: 25487016 DOI: 10.1007/978-3-319-11731-7_3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Hsp70/Hsp90 organising protein (Hop), also known as stress-inducible protein 1 (STI1), has received considerable attention for diverse cellular functions in both healthy and diseased states. There is extensive evidence that intracellular Hop is a co-chaperone of the major chaperones Hsp70 and Hsp90, playing an important role in the productive folding of Hsp90 client proteins. Consequently, Hop is implicated in a number of key signalling pathways, including aberrant pathways leading to cancer. However, Hop is also secreted and it is now well established that Hop also serves as a receptor for the prion protein, PrP(C). The intracellular and extracellular forms of Hop most likely represent two different isoforms, although the molecular determinants of these divergent functions are yet to be identified. There is also a growing body of research that reports the involvement of Hop in cellular activities that appear independent of either chaperones or PrP(C). While Hop has been shown to have various cellular functions, its biological function remains elusive. However, recent knockout studies in mammals suggest that Hop has an important role in embryonic development. This review provides a critical overview of the latest molecular, cellular and biological research on Hop, critically evaluating its function in healthy systems and how this function is adapted in diseases states.
Collapse
Affiliation(s)
- Swati Baindur-Hudson
- College of Health and Biomedicine, Victoria University, VIC 8001, Melbourne, Australia,
| | | | | |
Collapse
|
63
|
Haslene-Hox H, Oveland E, Woie K, Salvesen HB, Tenstad O, Wiig H. Distribution volumes of macromolecules in human ovarian and endometrial cancers--effects of extracellular matrix structure. Am J Physiol Heart Circ Physiol 2014; 308:H18-28. [PMID: 25380817 DOI: 10.1152/ajpheart.00672.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Elements of the extracellular matrix (ECM), notably collagen and glucosaminoglycans, will restrict part of the space available for soluble macromolecules simply because the molecules cannot occupy the same space. This phenomenon may influence macromolecular drug uptake. To study the influence of steric and charge effects of the ECM on the distribution volumes of macromolecules in human healthy and malignant gynecologic tissues we used as probes 15 abundant plasma proteins quantified by high-resolution mass spectrometry. The available distribution volume (VA) of albumin was increased in ovarian carcinoma compared with healthy ovarian tissue. Furthermore, VA of plasma proteins between 40 and 190 kDa decreased with size for endometrial carcinoma and healthy ovarian tissue, but was independent of molecular weight for the ovarian carcinomas. An effect of charge on distribution volume was only found in healthy ovaries, which had lower hydration and high collagen content, indicating that a condensed interstitium increases the influence of negative charges. A number of earlier suggested biomarker candidates were detected in increased amounts in malignant tissue, e.g., stathmin and spindlin-1, showing that interstitial fluid, even when unfractionated, can be a valuable source for tissue-specific proteins. We demonstrate that the distribution of abundant plasma proteins in the interstitium can be elucidated by mass spectrometry methods and depends markedly on hydration and ECM structure. Our data can be used in modeling of drug uptake, and give indications on ECM components to be targeted to increase the uptake of macromolecular substances.
Collapse
Affiliation(s)
| | - Eystein Oveland
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kathrine Woie
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway; and
| | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway; and Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway;
| |
Collapse
|
64
|
Kao C, Chao A, Tsai CL, Chuang WC, Huang WP, Chen GC, Lin CY, Wang TH, Wang HS, Lai CH. Bortezomib enhances cancer cell death by blocking the autophagic flux through stimulating ERK phosphorylation. Cell Death Dis 2014; 5:e1510. [PMID: 25375375 PMCID: PMC4260726 DOI: 10.1038/cddis.2014.468] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/10/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022]
Abstract
The antitumor activity of an inhibitor of 26S proteasome bortezomib (Velcade) has been observed in various malignancies, including colon cancer, prostate cancer, breast cancer, and ovarian cancer. Bortezomib has been proposed to stimulate autophagy, but scientific observations did not always support this. Interactions between ERK activity and autophagy are complex and not completely clear. Autophagy proteins have recently been shown to regulate the functions of ERK, and ERK activation has been found to induce autophagy. On the other hand, sustained activation of ERK has also been shown to inhibit the maturation step of the autophagy process. In this study, we sought to identify the mechanism of autophagy regulation in cancer cells treated with bortezomib. Our results indicate that bortezomib blocked the autophagic flux without inhibiting the fusion of the autophagosome and lysosome. In ovarian cancer, as well as endometrial cancer and hepatocellular carcinoma cells, bortezomib inhibited protein degradation in lysosomes by suppressing cathepsins, which requires the participation of ERK phosphorylation, but not JNK or p38. Our findings that ERK phosphorylation reduced cathepsins further explain how ERK phosphorylation inhibits the autophagic flux. In conclusion, bortezomib may induce ERK phosphorylation to suppress cathepsin B and inhibit the catalytic process of autophagy in ovarian cancer and other solid tumors. The inhibition of cisplatin-induced autophagy by bortezomib can enhance chemotherapy efficacy in ovarian cancer. As we also found that bortezomib blocks the autophagic flux in other cancers, the synergistic cytotoxic effect of bortezomib by abolishing chemotherapy-related autophagy may help us develop strategies of combination therapies for multiple cancers.
Collapse
Affiliation(s)
- C Kao
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - A Chao
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - C-L Tsai
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - W-C Chuang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - W-P Huang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - G-C Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - C-Y Lin
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - T-H Wang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan [3] Genomic Medicine Research Core Laboratory, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [4] School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - H-S Wang
- 1] Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan [2] Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - C-H Lai
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
65
|
Comparative proteomic analysis of fibrosarcoma and skin fibroblast cell lines. Tumour Biol 2014; 36:561-7. [DOI: 10.1007/s13277-014-2672-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/23/2014] [Indexed: 12/21/2022] Open
|
66
|
Disruption of prion protein-HOP engagement impairs glioblastoma growth and cognitive decline and improves overall survival. Oncogene 2014; 34:3305-14. [PMID: 25151961 DOI: 10.1038/onc.2014.261] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022]
Abstract
Glioblastomas (GBMs) are resistant to current therapy protocols and identification of molecules that target these tumors is crucial. Interaction of secreted heat-shock protein 70 (Hsp70)-Hsp90-organizing protein (HOP) with cellular prion protein (PrP(C)) triggers a large number of trophic effects in the nervous system. We found that both PrP(C) and HOP are highly expressed in human GBM samples relative to non-tumoral tissue or astrocytoma grades I-III. High levels of PrP(C) and HOP were associated with greater GBM proliferation and lower patient survival. HOP-PrP(C) binding increased GBM proliferation in vitro via phosphatidylinositide 3-kinase and extracellular-signal-regulated kinase pathways, and a HOP peptide mimicking the PrP(C) binding site (HOP230-245) abrogates this effect. PrP(C) knockdown impaired tumor growth and increased survival of mice with tumors. In mice, intratumor delivery of HOP230-245 peptide impaired proliferation and promoted apoptosis of GBM cells. In addition, treatment with HOP230-245 peptide inhibited tumor growth, maintained cognitive performance and improved survival. Thus, together, the present results indicate that interfering with PrP(C)-HOP engagement is a promising approach for GBM therapy.
Collapse
|
67
|
Carvalho da Fonseca AC, Wang H, Fan H, Chen X, Zhang I, Zhang L, Lima FRS, Badie B. Increased expression of stress inducible protein 1 in glioma-associated microglia/macrophages. J Neuroimmunol 2014; 274:71-7. [PMID: 25042352 DOI: 10.1016/j.jneuroim.2014.06.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/25/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
Factors released by glioma-associated microglia/macrophages (GAMs) play an important role in the growth and infiltration of tumors. We have previously demonstrated that the co-chaperone stress-inducible protein 1 (STI1) secreted by microglia promotes proliferation and migration of human glioblastoma (GBM) cell lines in vitro. In the present study, in order to investigate the role of STI1 in a physiological context, we used a glioma model to evaluate STI1 expression in vivo. Here, we demonstrate that STI1 expression in both the tumor and in the infiltrating GAMs and lymphocytes significantly increased with tumor progression. Interestingly, high expression of STI1 was observed in macrophages and lymphocytes that infiltrated brain tumors, whereas STI1 expression in the circulating blood monocytes and lymphocytes remained unchanged. Our results correlate, for the first time, the expression of STI1 and glioma progression, and suggest that STI1 expression in GAMs and infiltrating lymphocytes is modulated by the brain tumor microenvironment.
Collapse
Affiliation(s)
| | - Huaqing Wang
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Haitao Fan
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Xuebo Chen
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, PR China
| | - Ian Zhang
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States
| | - Leying Zhang
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States
| | - Flavia Regina Souza Lima
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Behnam Badie
- Division of Neurosurgery, Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute, Duarte, CA 91010, United States.
| |
Collapse
|
68
|
Lin CY, Chao A, Wang TH, Hsueh S, Lee YS, Wu TI, Chao AS, Huang HJ, Chou HH, Chang TC, Lai CH. A dual tyrosine kinase inhibitor lapatinib suppresses overexpression of matrix metallopeptidase 1 (MMP1) in endometrial cancer. J Mol Med (Berl) 2014; 92:969-81. [PMID: 24820961 DOI: 10.1007/s00109-014-1163-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/08/2014] [Accepted: 05/06/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED Endometrial cancers have been recently molecularly characterized; amplifications of human epidermal growth factor receptor 2 (HER2) were seen in 25 % of the serous-like tumors, and mutations in the PI(3)K/AKT pathways were seen in 93 % of endometrioid tumors. These new findings about endometrial cancer suggest a potential for targeted therapy with lapatinib, a dual inhibitor of epidermal growth factor receptor and HER2 tyrosine kinases. However, the clinical efficacy of lapatinib in phase II clinical trials for the treatment of endometrial cancers was only minimal. In this study, we investigated the signaling changes induced by lapatinib in endometrial cancer, which may improve its therapeutic efficacy in molecularly selected patient groups. We identified one of the final molecular targets of lapatinib to be interstitial collagenase, matrix metallopeptidase 1 (MMP1). Lapatinib suppresses MMP1 through EGFR and HER2, and their downstream ERK and AKT signaling pathways. We also found that the activating protein-1 binding site of MMP1 promoter is required for its transcriptional activation, which may be unique for endometrial cancers. Our results also showed that forced expression of active ERK or active AKT mutants rescued MMP1 expression from lapatinib suppression, further suggesting the importance of molecular selection to find appropriate patients with endometrial cancer for future clinical trials with any targeted therapies. KEY MESSAGE MMP1 expression was high in tissues and sera in patients with endometrial cancer. Lapatinib inhibited MMP1 via both HER2 and EGFR signaling pathways. Both AKT and ERK need to be inhibited for efficient MMP1 suppression by lapatinib. Activating protein-1 (AP-1) binding site of MMP1 promoter is uniquely required for MMP1 activation in endometrial cancer. Suppression of both c-fos and c-Jun bound to AP1 binding site is required for lapatinib inhibition.
Collapse
Affiliation(s)
- Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Van Simaeys D, Turek D, Champanhac C, Vaizer J, Sefah K, Zhen J, Sutphen R, Tan W. Identification of cell membrane protein stress-induced phosphoprotein 1 as a potential ovarian cancer biomarker using aptamers selected by cell systematic evolution of ligands by exponential enrichment. Anal Chem 2014; 86:4521-7. [PMID: 24654750 PMCID: PMC4018121 DOI: 10.1021/ac500466x] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 03/21/2014] [Indexed: 02/01/2023]
Abstract
In this paper, we describe the elucidation of the target of an aptamer against ovarian cancer previously obtained by cell-SELEX (SELEX = systematic evolution of ligands by exponential enrichment). The target's identity, stress-induced phosphoprotein 1 (STIP1), was determined by mass spectrometry and validated by flow cytometry, using siRNA silencing and protein blotting. Initial oncologic studies show that the aptamer inhibits cell invasion, indicating that STIP1, which is currently under investigation as a potential biomarker for ovarian cancer, plays a critical role in this process. These results serve as an excellent example of how protein target identification of aptamers obtained by cell-SELEX can serve as a means to identify promising biomarker candidates and can promote the development of aptamers as a new drug class to block important oncological processes.
Collapse
Affiliation(s)
- Dimitri Van Simaeys
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Diane Turek
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Carole Champanhac
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Julia Vaizer
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Kwame Sefah
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Jing Zhen
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
- Molecular
Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing
and Chemometrics, Colleges of Chemistry and Chemical Engineering and
of Biology, Collaborative Innovation Center for Molecular Engineering
and Theranostics, Hunan University, Changsha 410082, China
| | - Rebecca Sutphen
- Morsani
School of Medicine, University of South
Florida, Tampa, Florida 33612, United
States
| | - Weihong Tan
- Center
for Research at Bio/Nano Interface, Departments of Chemistry and of
Physiology and Functional Genomics, Shands Cancer Center, UF Genetics
Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
- Molecular
Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing
and Chemometrics, Colleges of Chemistry and Chemical Engineering and
of Biology, Collaborative Innovation Center for Molecular Engineering
and Theranostics, Hunan University, Changsha 410082, China
| |
Collapse
|
70
|
STI1 antagonizes cytoskeleton collapse mediated by small GTPase Rnd1 and regulates neurite growth. Exp Cell Res 2014; 324:84-91. [PMID: 24690281 DOI: 10.1016/j.yexcr.2014.03.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/19/2014] [Accepted: 03/22/2014] [Indexed: 01/10/2023]
Abstract
Rnd proteins comprise a branch of the Rho family of small GTP-binding proteins, which have been implicated in rearrangements of the actin cytoskeleton and microtubule dynamics. Particularly in the nervous system, Rnd family proteins regulate neurite formation, dendrite development and axonal branching. A secreted form of the co-chaperone Stress-Inducible Protein 1 (STI1) has been described as a prion protein partner that is involved in several processes of the nervous system, such as neurite outgrowth, neuroprotection, astrocyte development, and the self-renewal of neural progenitor cells. We show that cytoplasmic STI1 directly interacts with the GTPase Rnd1. This interaction is specific for the Rnd1 member of the Rnd family. In the COS collapse assay, overexpression of STI1 prevents Rnd1-plexin-A1-mediated cytoskeleton retraction. In PC-12 cells, overexpression of STI1 enhances neurite outgrowth in cellular processes initially established by Rnd1. Therefore, we propose that STI1 participates in Rnd1-induced signal transduction pathways that are involved in the dynamics of the actin cytoskeleton.
Collapse
|
71
|
Cho H, Kim S, Shin HY, Chung EJ, Kitano H, Hyon Park J, Park L, Chung JY, Hewitt SM, Kim JH. Expression of stress-induced phosphoprotein1 (STIP1) is associated with tumor progression and poor prognosis in epithelial ovarian cancer. Genes Chromosomes Cancer 2014; 53:277-88. [PMID: 24488757 DOI: 10.1002/gcc.22136] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 11/11/2022] Open
Abstract
Stress-induced phosphoprotein1 (STIP1) is a candidate biomarker in epithelial ovarian cancer (EOC). In this study, we investigated in detail the expression of STIP1, as well as its functions, in EOC. STIP1 expression was assessed by immunohistochemistry (IHC) and the results were compared with clinicopathologic factors, including survival data. The effects of STIP1 gene silencing via small interfering RNA (siRNA) were examined in EOC cells and a xenograft model. The expression of STIP1 protein in EOC was significantly higher than in the other study groups (P < 0.001), and this increase of expression was significantly associated with tumor stage (P = 0.005), tumor grade (P = 0.029), and lymph node metastasis (P = 0.020). In multivariate analysis, overall survival in EOC was significantly shorter in cases with high STIP1 expression (HR = 2.78 [1.01-7.63], P = 0.047). STIP1 silencing in EOC cells resulted in inhibition of cell proliferation and invasion. In addition, in vivo experiments using STIP1 siRNA clearly showed a strong inhibition of tumor growth and a modulation of expression of prosurvival and apoptotic genes, further suggesting that STIP1 silencing can prevent cell proliferation and invasion. In conclusion, increased STIP1 expression is associated with poor survival outcome in EOC, and STIP1 may represent a useful therapeutic target in EOC patients.
Collapse
Affiliation(s)
- Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Karsani SA, Saihen NA, Zain RB, Cheong SC, Abdul Rahman M. Comparative proteomics analysis of oral cancer cell lines: identification of cancer associated proteins. Proteome Sci 2014; 12:3. [PMID: 24422745 PMCID: PMC3974152 DOI: 10.1186/1477-5956-12-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 01/07/2014] [Indexed: 12/03/2022] Open
Abstract
Background A limiting factor in performing proteomics analysis on cancerous cells is the difficulty in obtaining sufficient amounts of starting material. Cell lines can be used as a simplified model system for studying changes that accompany tumorigenesis. This study used two-dimensional gel electrophoresis (2DE) to compare the whole cell proteome of oral cancer cell lines vs normal cells in an attempt to identify cancer associated proteins. Results Three primary cell cultures of normal cells with a limited lifespan without hTERT immortalization have been successfully established. 2DE was used to compare the whole cell proteome of these cells with that of three oral cancer cell lines. Twenty four protein spots were found to have changed in abundance. MALDI TOF/TOF was then used to determine the identity of these proteins. Identified proteins were classified into seven functional categories – structural proteins, enzymes, regulatory proteins, chaperones and others. IPA core analysis predicted that 18 proteins were related to cancer with involvements in hyperplasia, metastasis, invasion, growth and tumorigenesis. The mRNA expressions of two proteins – 14-3-3 protein sigma and Stress-induced-phosphoprotein 1 – were found to correlate with the corresponding proteins’ abundance. Conclusions The outcome of this analysis demonstrated that a comparative study of whole cell proteome of cancer versus normal cell lines can be used to identify cancer associated proteins.
Collapse
Affiliation(s)
- Saiful Anuar Karsani
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | | | | | | | | |
Collapse
|
73
|
Cheng SW, Tsai HW, Lin YJ, Cheng PN, Chang YC, Yen CJ, Huang HP, Chuang YP, Chang TT, Lee CT, Chao A, Chou CY, Chan SH, Chow NH, Ho CL. Lin28B is an oncofetal circulating cancer stem cell-like marker associated with recurrence of hepatocellular carcinoma. PLoS One 2013; 8:e80053. [PMID: 24244607 PMCID: PMC3828221 DOI: 10.1371/journal.pone.0080053] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 09/30/2013] [Indexed: 12/21/2022] Open
Abstract
By using an expressed sequence tag bioinformatic algorithm, we identified that Lin28 homolog B (Lin28B) may have an oncofetal expression pattern which may facilitate detecting cancer cells in adults. It is also reported to be a potential marker for cancer stem cells. Therefore, we sought to verify oncofetal-stemness characters of Lin28B and test its potential as a circulating cancer stem cell-like marker in adult HCC patients. Lin28B mRNA was examined in a panel of fetal tissue, adult tissue and tumors. Lin28B was over-expressed or knocked down in HepG2 cells to evaluate its potential as a stem cell-like marker. RT-qPCR for Lin28B was performed in the peripheral blood mononuclear cells from patients with HCC receiving surgery (n=96) and non-HCC controls (n=60) and analyzed its clinical significance. Lin28B showed an oncofetal expression pattern. Its overexpression could upregulate stemness markers (OCT4, Nanog and SOX2) and enhance tumorsphere formation in vitro. Lin28B knockdown had opposite effects. Circulating Lin28B was detected in peripheral blood mononuclear cells in 3 cases (5%) of non-HCC controls and 32 cases (33.3%) of HCC patients. In HCC patients, circulating Lin28B was associated with high tumor grade (P=0.046), large size (P=0.005), high AJCC stage (P=0.044) and BCLC stage (P=0.017). Circulating Lin28B was significantly associated with decreased recurrence-free survival (P<0.001). Circulating Lin28B separated early stage HCC into 2 recurrence-free survival curves (P=0.003). In multivariate analysis, circulating Lin28B was an independent variable associated with early recurrence (P=0.045) and recurrence in early stage HCC (P=0.006). In conclusion, the oncofetal gene Lin28B is a potential oncofetal cancer-stem-cell-like circulating tumor cell marker that correlates with HCC recurrence after hepatectomy. Circulating Lin28B could refine early AJCC stages. Our finding supports the possible use of a TNMC (C for circulating tumor cells) staging system in HCC.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Female
- Gene Expression Regulation, Neoplastic
- Hep G2 Cells
- Hepatectomy
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- Male
- Middle Aged
- Nanog Homeobox Protein
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/surgery
- Neoplasm Staging
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Octamer Transcription Factor-3/genetics
- Octamer Transcription Factor-3/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- SOXB1 Transcription Factors/genetics
- SOXB1 Transcription Factors/metabolism
- Survival Analysis
Collapse
Affiliation(s)
- Shu-Wen Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Gromov P, Gromova I, Olsen CJ, Timmermans-Wielenga V, Talman ML, Serizawa RR, Moreira JM. Tumor interstitial fluid — A treasure trove of cancer biomarkers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2259-70. [DOI: 10.1016/j.bbapap.2013.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 12/11/2022]
|
75
|
Yuan MH, Zhou RS, She B, Xu HF, Wang JY, Wei LX. Expression and clinical significance of STIP1 in papillary thyroid carcinoma. Tumour Biol 2013; 35:2391-5. [DOI: 10.1007/s13277-013-1316-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/14/2013] [Indexed: 12/21/2022] Open
|
76
|
Estradiol and tamoxifen induce cell migration through GPR30 and activation of focal adhesion kinase (FAK) in endometrial cancers with low or without nuclear estrogen receptor α (ERα). PLoS One 2013; 8:e72999. [PMID: 24039841 PMCID: PMC3767783 DOI: 10.1371/journal.pone.0072999] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022] Open
Abstract
Estrogens and tamoxifen (an antiestrogen) exert their actions by activation of estrogen receptor (ER) through genomic and non-genomic mechanisms and are implicated in the development of endometrial cancer. Previous reports have demonstrated that estradiol and tamoxifen induce proliferation of human endometrial cancer cells through GPR30 (non-genomic ER) signaling pathway. Herein, we demonstrate that phosphorylation of focal adhesion kinase (FAK) is involved in cell migration induced by estradiol, tamoxifen and G1 (a GPR30 agonist) through the transmembrane ER (GPR30) in endometrial cancer cell lines with or without ERα (Ishikawa and RL95-2). Additionally, the GPR30-mediated cell migration was further abolished by administration of either specific RNA interference targeting GPR30 or an FAK inhibitor. Moreover, we have validated that the signaling between GPR30 and phosphorylated FAK is indeed mediated by the EGFR/PI3K/ERK pathway. Clinically, a significant correlation between levels of GPR30 and phophorylated FAK (pFAK) observed in human endometrial cancer tissues with low or without ERα further suggested that estrogen-induced phosphorylation of FAK and cell migration were most likely triggered by GPR30 activation. These results provided new insights for understanding the pathophysiological functions of GPR30 in human endometrial cancers.
Collapse
|
77
|
Lee J, Cho S, Lee J, Ryu H, Park J, Lim S, Oh B, Lee C, Huang W, Busnaina A, Lee H. Wafer-scale nanowell array patterning based electrochemical impedimetric immunosensor. J Biotechnol 2013; 168:584-8. [PMID: 24013070 DOI: 10.1016/j.jbiotec.2013.08.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 08/09/2013] [Accepted: 08/22/2013] [Indexed: 10/26/2022]
Abstract
We have reported that nanowell array (NWA) can enhance electrochemical detection of molecular binding events by controlling the binding sites of the captured molecules. Using NWA biosensor based amperometric analysis, we have detected biological macromolecules such as DNA, protein or aptamers at low concentrations. In this research, we developed an impedimetric immunosensor based on wafer-scale NWA for electrochemical detection of stress-induced-phosphoprotein-1 (STIP-1). In order to develop NWA sensor through the cost-effective combination of high-throughput nanopattern, the NWA electrode was fabricated on Si wafer by krypton-fluoride (KrF) stepper semiconductor process. Finally, 12,500,000 ea nanowell with a 500 nm diameter was fabricated on 4 mm × 2 mm substrate. Next, by using these electrodes, we measured impedance to quantify antigen binding to the immunoaffinity layer. The limit of detection (LOD) of the NWA was improved about 100-fold compared to milli-sized electrodes (4 mm × 2 mm) without an NWA. These results suggest that wafer-scale NWA immunosensor will be useful for biosensing applications because their interface response is appropriate for detecting molecular binding events.
Collapse
Affiliation(s)
- JuKyung Lee
- Department of Mechanical and Industrial Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Hajj GNM, Arantes CP, Dias MVS, Roffé M, Costa-Silva B, Lopes MH, Porto-Carreiro I, Rabachini T, Lima FR, Beraldo FH, Prado MMA, Linden R, Martins VR. The unconventional secretion of stress-inducible protein 1 by a heterogeneous population of extracellular vesicles. Cell Mol Life Sci 2013; 70:3211-27. [PMID: 23543276 PMCID: PMC11113396 DOI: 10.1007/s00018-013-1328-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/15/2013] [Accepted: 03/18/2013] [Indexed: 01/03/2023]
Abstract
The co-chaperone stress-inducible protein 1 (STI1) is released by astrocytes, and has important neurotrophic properties upon binding to prion protein (PrP(C)). However, STI1 lacks a signal peptide and pharmacological approaches pointed that it does not follow a classical secretion mechanism. Ultracentrifugation, size exclusion chromatography, electron microscopy, vesicle labeling, and particle tracking analysis were used to identify three major types of extracellular vesicles (EVs) released from astrocytes with sizes ranging from 20-50, 100-200, and 300-400 nm. These EVs carry STI1 and present many exosomal markers, even though only a subpopulation had the typical exosomal morphology. The only protein, from those evaluated here, present exclusively in vesicles that have exosomal morphology was PrP(C). STI1 partially co-localized with Rab5 and Rab7 in endosomal compartments, and a dominant-negative for vacuolar protein sorting 4A (VPS4A), required for formation of multivesicular bodies (MVBs), impaired EV and STI1 release. Flow cytometry and PK digestion demonstrated that STI1 localized to the outer leaflet of EVs, and its association with EVs greatly increased STI1 activity upon PrP(C)-dependent neuronal signaling. These results indicate that astrocytes secrete a diverse population of EVs derived from MVBs that contain STI1 and suggest that the interaction between EVs and neuronal surface components enhances STI1-PrP(C) signaling.
Collapse
Affiliation(s)
- Glaucia N. M. Hajj
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Camila P. Arantes
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Marcos Vinicios Salles Dias
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Martín Roffé
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Bruno Costa-Silva
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Marilene H. Lopes
- Department of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isabel Porto-Carreiro
- Instituto de Biofisica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Flávia R. Lima
- Instituto de Ciências Biomédicas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávio H. Beraldo
- Department of Anatomy and Cell Biology and Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Marco M. A. Prado
- Department of Anatomy and Cell Biology and Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Rafael Linden
- Instituto de Biofisica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vilma R. Martins
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| |
Collapse
|
79
|
Olsson N, Carlsson P, James P, Hansson K, Waldemarson S, Malmström P, Fernö M, Ryden L, Wingren C, Borrebaeck CAK. Grading breast cancer tissues using molecular portraits. Mol Cell Proteomics 2013; 12:3612-23. [PMID: 23982162 PMCID: PMC3861711 DOI: 10.1074/mcp.m113.030379] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Tumor progression and prognosis in breast cancer patients are difficult to assess using current clinical and laboratory parameters, where a pathological grading is indicative of tumor aggressiveness. This grading is based on assessments of nuclear grade, tubule formation, and mitotic rate. We report here the first protein signatures associated with histological grades of breast cancer, determined using a novel affinity proteomics approach. We profiled 52 breast cancer tissue samples by combining nine antibodies and label-free LC-MS/MS, which generated detailed quantified proteomic maps representing 1,388 proteins. The results showed that we could define in-depth molecular portraits of histologically graded breast cancer tumors. Consequently, a 49-plex candidate tissue protein signature was defined that discriminated between histological grades 1, 2, and 3 of breast cancer tumors with high accuracy. Highly biologically relevant proteins were identified, and the differentially expressed proteins indicated further support for the current hypothesis regarding remodeling of the tumor microenvironment during tumor progression. The protein signature was corroborated using meta-analysis of transcriptional profiling data from an independent patient cohort. In addition, the potential for using the markers to estimate the likelihood of long-term metastasis-free survival was also indicated. Taken together, these molecular portraits could pave the way for improved classification and prognostication of breast cancer.
Collapse
Affiliation(s)
- Niclas Olsson
- Department of Immunotechnology and CREATE HEALTH, Lund University, Medicon Village, SE-223 81 Lund, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Soares IN, Caetano FA, Pinder J, Rodrigues BR, Beraldo FH, Ostapchenko VG, Durette C, Pereira GS, Lopes MH, Queiroz-Hazarbassanov N, Cunha IW, Sanematsu PI, Suzuki S, Bleggi-Torres LF, Schild-Poulter C, Thibault P, Dellaire G, Martins VR, Prado VF, Prado MAM. Regulation of stress-inducible phosphoprotein 1 nuclear retention by protein inhibitor of activated STAT PIAS1. Mol Cell Proteomics 2013; 12:3253-70. [PMID: 23938469 DOI: 10.1074/mcp.m113.031005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Stress-inducible phosphoprotein 1 (STI1), a cochaperone for Hsp90, has been shown to regulate multiple pathways in astrocytes, but its contributions to cellular stress responses are not fully understood. We show that in response to irradiation-mediated DNA damage stress STI1 accumulates in the nucleus of astrocytes. Also, STI1 haploinsufficiency decreases astrocyte survival after irradiation. Using yeast two-hybrid screenings we identified several nuclear proteins as STI1 interactors. Overexpression of one of these interactors, PIAS1, seems to be specifically involved in STI1 nuclear retention and in directing STI1 and Hsp90 to specific sub-nuclear regions. PIAS1 and STI1 co-immunoprecipitate and PIAS1 can function as an E3 SUMO ligase for STI. Using mass spectrometry we identified five SUMOylation sites in STI1. A STI1 mutant lacking these five sites is not SUMOylated, but still accumulates in the nucleus in response to increased expression of PIAS1, suggesting the possibility that a direct interaction with PIAS1 could be responsible for STI1 nuclear retention. To test this possibility, we mapped the interaction sites between PIAS1 and STI1 using yeast-two hybrid assays and surface plasmon resonance and found that a large domain in the N-terminal region of STI1 interacts with high affinity with amino acids 450-480 of PIAS1. Knockdown of PIAS1 in astrocytes impairs the accumulation of nuclear STI1 in response to irradiation. Moreover, a PIAS1 mutant lacking the STI1 binding site is unable to increase STI1 nuclear retention. Interestingly, in human glioblastoma multiforme PIAS1 expression is increased and we found a significant correlation between increased PIAS1 expression and STI1 nuclear localization. These experiments provide evidence that direct interaction between STI1 and PIAS1 is involved in the accumulation of nuclear STI1. This retention mechanism could facilitate nuclear chaperone activity.
Collapse
Affiliation(s)
- Iaci N Soares
- Robarts Research Institute, The University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Chao A, Lee LY, Hsueh C, Lin CY, Tsai CL, Chao AS, Lin CT, Chou HH, Chang TC, Wang TH. Immunohistological analysis of stress-induced phosphoprotein 1 in ovarian cancer patients with low serum cancer antigen 125 levels. Taiwan J Obstet Gynecol 2013; 52:185-91. [DOI: 10.1016/j.tjog.2013.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2013] [Indexed: 10/26/2022] Open
|
82
|
Beraldo FH, Soares IN, Goncalves DF, Fan J, Thomas AA, Santos TG, Mohammad AH, Roffé M, Calder MD, Nikolova S, Hajj GN, Guimaraes AL, Massensini AR, Welch I, Betts DH, Gros R, Drangova M, Watson AJ, Bartha R, Prado VF, Martins VR, Prado MAM. Stress-inducible phosphoprotein 1 has unique cochaperone activity during development and regulates cellular response to ischemia via the prion protein. FASEB J 2013; 27:3594-607. [PMID: 23729591 DOI: 10.1096/fj.13-232280] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stress-inducible phosphoprotein 1 (STI1) is part of the chaperone machinery, but it also functions as an extracellular ligand for the prion protein. However, the physiological relevance of these STI1 activities in vivo is unknown. Here, we show that in the absence of embryonic STI1, several Hsp90 client proteins are decreased by 50%, although Hsp90 levels are unaffected. Mutant STI1 mice showed increased caspase-3 activation and 50% impairment in cellular proliferation. Moreover, placental disruption and lack of cellular viability were linked to embryonic death by E10.5 in STI1-mutant mice. Rescue of embryonic lethality in these mutants, by transgenic expression of the STI1 gene, supported a unique role for STI1 during embryonic development. The response of STI1 haploinsufficient mice to cellular stress seemed compromised, and mutant mice showed increased vulnerability to ischemic insult. At the cellular level, ischemia increased the secretion of STI1 from wild-type astrocytes by 3-fold, whereas STI1 haploinsufficient mice secreted half as much STI1. Interesting, extracellular STI1 prevented ischemia-mediated neuronal death in a prion protein-dependent way. Our study reveals essential roles for intracellular and extracellular STI1 in cellular resilience.
Collapse
Affiliation(s)
- Flavio H Beraldo
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Gupta MK, Polisetty RV, Ramamoorthy K, Tiwary S, Kaur N, Uppin MS, Shiras A, Sirdeshmukh R. Secretome analysis of Glioblastoma cell line--HNGC-2. MOLECULAR BIOSYSTEMS 2013; 9:1390-400. [PMID: 23483059 DOI: 10.1039/c3mb25383j] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of primary malignant tumor of the central nervous system. We have carried out a deep analysis of the secretome of a rapidly proliferating and tumorigenic cell line HNGC-2, representing GBM, in an effort to identify proteins, which may be targeted in the plasma of GBM patients as markers for diagnosis and disease surveillance. Prefractionation of the proteins from the conditioned medium of HNGC-2 cells in SDS gels followed by LC-MS/MS analysis using an ESI-IT mass spectrometer (LTQ) led to a total of 996 protein identifications with ≥2 peptides each. Of them, 664 proteins were observed in the transcriptome of HNGC-2 cells. The dataset of 996 proteins was mapped to important functional groups, such as cellular assembly and organisation, DNA recombination and repair, and other classes. Actin cytoskeleton signalling, phosphatidyl inositol 3 kinase (PI3K/AKT) and integrin linked kinase (ILK) signalling pathways were seen as enriched pathways. Comparisons with the published secretome of cell lines from 12 different cancers, including GBM, revealed that 348 proteins shared a commonality with a secretome of at least one other cell line, 321 of which were found to contain signal sequences or transmembrane domains and 335 could be linked to a plasma membrane or extracellular localization. Through intergration of this data we arrived at a non-redundant list of 597 protein identifications with the potential for secretion either by classical secretory pathways or by non-secretory processes; 233 of them have been detected in cerebrospinal fluid or plasma as per the published literature, and 172 have been implicated in GBM or other cancers. The HNGC-2 secretome dataset could serve as a useful resource for designing a targeted investigation of GBM biomarkers in plasma.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, 500007, India
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Microproteomics by liquid extraction surface analysis: Application to FFPE tissue to study the fimbria region of tubo-ovarian cancer. Proteomics Clin Appl 2013; 7:234-40. [DOI: 10.1002/prca.201200070] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/26/2012] [Accepted: 12/17/2012] [Indexed: 11/07/2022]
|
85
|
Chao A, Lai CH, Tsai CL, Hsueh S, Hsueh C, Lin CY, Chou HH, Lin YJ, Chen HW, Chang TC, Wang TH. Tumor stress-induced phosphoprotein1 (STIP1) as a prognostic biomarker in ovarian cancer. PLoS One 2013; 8:e57084. [PMID: 23468915 PMCID: PMC3584135 DOI: 10.1371/journal.pone.0057084] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 01/16/2013] [Indexed: 12/20/2022] Open
Abstract
Stress-induced phosphoprotein 1 (STIP1) has been recently identified as a released biomarker in human ovarian cancer. In addition, STIP1 secreted by human ovarian cancer cells has been shown to promote tumor cell proliferation by binding to ALK2 (activin A receptor, type II-like kinase 2) and activating the SMAD-ID3 signaling pathways. In this study, a total of 330 ovarian cancer tumor samples were evaluated for STIP1 expression by immunohistochemistry and analyzed for a possible correlation with patient characteristics and survival. The quantification of immunoreactivity was accomplished by applying an immunohistochemical scoring system (histoscore). Patients with high-level STIP1 expression (histoscore ≥169) had a significantly worse survival (high STIP1, mean survival time = 76 months; low STIP1, mean survival time = 112 months; P<0.0001). Moreover, STIP1 histoscores were significantly higher in high-grade tumors (grade 3) than in low-grade (grade 1–2) malignancies (P<0.0001), suggesting that STIP1 may be a proxy for tumor aggressiveness. The results of multivariable analysis revealed that high STIP1 histoscores, advanced stages, histologic types, and the presence of residual disease (≥2 cm) were independent predictors of poor prognosis. The addition of STIP1 histoscores improved the prediction of overall and progression-free survival rates in the multivariable Cox proportional hazard model. The treatment of ovarian cancer cells with recombinant STIP1 stimulated cell proliferation and migration, but co-treatment with anti-STIP1 antibodies abrogated this effect. Our findings suggest that STIP1 expression may be related to prognosis and that the STIP1 pathway may represent a novel therapeutic target for human ovarian cancer.
Collapse
Affiliation(s)
- Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- * E-mail: (AC); (THW)
| | - Chyong-Huey Lai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chia-Lung Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Swei Hsueh
- Department of Clinical Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chuen Hsueh
- Department of Clinical Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Hung-Hsueh Chou
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yu-Jr Lin
- Biostatistical Center for Clinical Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsi-Wen Chen
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Chang Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Hao Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail: (AC); (THW)
| |
Collapse
|
86
|
Haslene-Hox H, Tenstad O, Wiig H. Interstitial fluid-a reflection of the tumor cell microenvironment and secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2336-46. [PMID: 23376185 DOI: 10.1016/j.bbapap.2013.01.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/27/2012] [Accepted: 01/24/2013] [Indexed: 02/07/2023]
Abstract
The interstitium or interstitial space describes the space outside the blood and lymphatic vessels. It contains two phases; the interstitial fluid (IF) and the extracellular matrix. In this review we focus on the interstitial fluid phase, which is the physical and biochemical microenvironment of the cells, and more specifically that of tumors. IF is created by transcapillary filtration and cleared by lymphatic vessels, and contains substances that are either produced and secreted locally, thus denoted secretome, or brought to the organ by the circulation. The structure of the interstitium is discussed briefly and moreover techniques for IF isolation focusing on those that are relevant for studies of the secretome. Accumulated data show that tumor IF is hypoxic and acidic compared with subcutaneous IF and plasma, and that there are gradients between IF and plasma giving information on where substances are produced and thereby reflecting the local microenvironment. We review recent data on the origin of tissue specific substances, challenges related to isolating a representative secretome and the use of this as a substrate for biomarker identification. Finally we perform a comparative analysis across human tumor types and techniques and show that there is great variation in the results obtained that may at least partially be due to the isolation method used. We conclude that when care is taken in isolation of substrate, analysis of the secretome may give valuable biological insight and result in identification of biomarker candidates. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
|
87
|
Zhang Y, Xu B, Liu Y, Yao H, Lu N, Li B, Gao J, Guo S, Han N, Qi J, Zhang K, Cheng S, Wang H, Zhang X, Xiao T, Wu L, Gao Y. The ovarian cancer-derived secretory/releasing proteome: A repertoire of tumor markers. Proteomics 2012; 12:1883-91. [PMID: 22623176 DOI: 10.1002/pmic.201100654] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy worldwide, and early detection of this disease using serum or plasma biomarkers may improve its clinical outcome. In the present study, a large scale protein database derived from ovarian cancer was created to enable tumor marker discovery. First, primary organ cultures were established with the tumor tissues and corresponding normal tissues obtained from six ovarian cancer patients, and the serum-free conditioned medium (CM) samples were collected for proteomic analysis. The total proteins from the CM sample were separated by SDS-PAGE, digested with trypsin and then analyzed by LC-MS/MS. Combining data from the tumor tissues and the normal tissues, 1129 proteins were identified in total, of which those categorized as "extracellular proteins" and "plasma membrane proteins" accounted for 21.4% and 16.9%, respectively. For validation, three secretory proteins (NID1, TIMP2, and VCAN) involved in "organ development"-associated subnetwork, showed significant differences between their levels in the circulating plasma samples from ovarian cancer patients and healthy women. In conclusion, this ovarian cancer-derived protein database provides a credible repertoire of potential biomarkers in blood for this malignant disease, and deserves mining further.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Cancer Institute (Hospital), Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Santos TG, Beraldo FH, Hajj GNM, Lopes MH, Roffe M, Lupinacci FCS, Ostapchenko VG, Prado VF, Prado MAM, Martins VR. Laminin-γ1 chain and stress inducible protein 1 synergistically mediate PrPC-dependent axonal growth via Ca2+ mobilization in dorsal root ganglia neurons. J Neurochem 2012; 124:210-23. [PMID: 23145988 DOI: 10.1111/jnc.12091] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 11/01/2012] [Accepted: 11/01/2012] [Indexed: 12/01/2022]
Abstract
Prion protein (PrP(C)) is a cell surface glycoprotein that is abundantly expressed in nervous system. The elucidation of the PrP(C) interactome network and its significance on neural physiology is crucial to understanding neurodegenerative events associated with prion and Alzheimer's diseases. PrP(C) co-opts stress inducible protein 1/alpha7 nicotinic acetylcholine receptor (STI1/α7nAChR) or laminin/Type I metabotropic glutamate receptors (mGluR1/5) to modulate hippocampal neuronal survival and differentiation. However, potential cross-talk between these protein complexes and their role in peripheral neurons has never been addressed. To explore this issue, we investigated PrP(C)-mediated axonogenesis in peripheral neurons in response to STI1 and laminin-γ1 chain-derived peptide (Ln-γ1). STI1 and Ln-γ1 promoted robust axonogenesis in wild-type neurons, whereas no effect was observed in neurons from PrP(C) -null mice. PrP(C) binding to Ln-γ1 or STI1 led to an increase in intracellular Ca(2+) levels via distinct mechanisms: STI1 promoted extracellular Ca(2+) influx, and Ln-γ1 released calcium from intracellular stores. Both effects depend on phospholipase C activation, which is modulated by mGluR1/5 for Ln-γ1, but depends on, C-type transient receptor potential (TRPC) channels rather than α7nAChR for STI1. Treatment of neurons with suboptimal concentrations of both ligands led to synergistic actions on PrP(C)-mediated calcium response and axonogenesis. This effect was likely mediated by simultaneous binding of the two ligands to PrP(C). These results suggest a role for PrP(C) as an organizer of diverse multiprotein complexes, triggering specific signaling pathways and promoting axonogenesis in the peripheral nervous system.
Collapse
Affiliation(s)
- Tiago G Santos
- International Research Center, A.C. Camargo Hospital, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Chao A, Lin CY, Tsai CL, Hsueh S, Lin YY, Lin CT, Chou HH, Wang TH, Lai CH, Wang HS. Estrogen stimulates the proliferation of human endometrial cancer cells by stabilizing nucleophosmin/B23 (NPM/B23). J Mol Med (Berl) 2012; 91:249-59. [PMID: 22926011 DOI: 10.1007/s00109-012-0950-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/06/2012] [Accepted: 08/16/2012] [Indexed: 11/30/2022]
Abstract
Unopposed estrogen exposure is an important factor in the tumorigenesis of endometrial cancer. Nucleophosmin/B23 (NPM/B23), a phosphoprotein that has pleiotropic functions in cells, plays an important role in various cancers. However, the regulatory role of NPM/B23 in estrogen signaling in endometrial cancer has not been explored. Here, we report that NPM/B23 was required for estrogen-induced endometrial proliferation, and the increase in NPM/B23 was estrogen receptor α-dependent. Furthermore, estrogen increased NPM/B23 protein levels by repressing its ubiquitination and subsequently stabilizing the protein. The overexpression of the alternate reading frame (ARF) suppressed the estrogen-induced increase in the NPM/B23 protein levels, indicating that ARF inhibited the observed estrogen-mediated NPM/B23 stabilization. Our results suggest that one of the effects of estrogen on endometrial proliferation is the suppression of the NPM/B23-ARF interaction and the subsequent increase in NPM/B23 protein levels. This novel characterization of NPM/B23 in estrogen-mediated cell proliferation may extend our understanding of the tumorigenesis of steroid hormone-related cancers.
Collapse
Affiliation(s)
- Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Linkou Medical Center, 5 Fushin Street, Guishan, Taoyuan, 333, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Secreted Stress-Induced Phosphoprotein 1 Activates the ALK2-SMAD Signaling Pathways and Promotes Cell Proliferation of Ovarian Cancer Cells. Cell Rep 2012; 2:283-93. [DOI: 10.1016/j.celrep.2012.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 06/22/2012] [Accepted: 07/06/2012] [Indexed: 11/22/2022] Open
|
91
|
Santos TG, Silva IR, Costa-Silva B, Lepique AP, Martins VR, Lopes MH. Enhanced neural progenitor/stem cells self-renewal via the interaction of stress-inducible protein 1 with the prion protein. Stem Cells 2011; 29:1126-36. [PMID: 21608082 DOI: 10.1002/stem.664] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prion protein (PrP(C) ), when associated with the secreted form of the stress-inducible protein 1 (STI1), plays an important role in neural survival, neuritogenesis, and memory formation. However, the role of the PrP(C) -STI1 complex in the physiology of neural progenitor/stem cells is unknown. In this article, we observed that neurospheres cultured from fetal forebrain of wild-type (Prnp(+/+) ) and PrP(C) -null (Prnp(0/0) ) mice were maintained for several passages without the loss of self-renewal or multipotentiality, as assessed by their continued capacity to generate neurons, astrocytes, and oligodendrocytes. The homogeneous expression and colocalization of STI1 and PrP(C) suggest that they may associate and function as a complex in neurosphere-derived stem cells. The formation of neurospheres from Prnp(0/0) mice was reduced significantly when compared with their wild-type counterparts. In addition, blockade of secreted STI1, and its cell surface ligand, PrP(C) , with specific antibodies, impaired Prnp(+/+) neurosphere formation without further impairing the formation of Prnp(0/0) neurospheres. Alternatively, neurosphere formation was enhanced by recombinant STI1 application in cells expressing PrP(C) but not in cells from Prnp(0/0) mice. The STI1-PrP(C) interaction was able to stimulate cell proliferation in the neurosphere-forming assay, while no effect on cell survival or the expression of neural markers was observed. These data suggest that the STI1-PrP(C) complex may play a critical role in neural progenitor/stem cells self-renewal via the modulation of cell proliferation, leading to the control of the stemness capacity of these cells during nervous system development.
Collapse
Affiliation(s)
- Tiago G Santos
- Department of Molecular and Cell Biology, International Center for Research and Education, Antonio Prudente Foundation, A. C. Camargo Hospital and National Institute for Translational Neuroscience (CNPq/MCT), São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
92
|
Teng PN, Bateman NW, Hood BL, Conrads TP. Advances in proximal fluid proteomics for disease biomarker discovery. J Proteome Res 2010; 9:6091-100. [PMID: 21028795 DOI: 10.1021/pr100904q] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although serum/plasma has been the preferred source for identification of disease biomarkers, these efforts have been met with little success, in large part due the relatively small number of highly abundant proteins that render the reliable detection of low abundant disease-related proteins challenging due to the expansive dynamic range of concentration of proteins in this sample. Proximal fluid, the fluid derived from the extracellular milieu of tissues, contains a large repertoire of shed and secreted proteins that are likely to be present at higher concentrations relative to plasma/serum. It is hypothesized that many, if not all, proximal fluid proteins exchange with peripheral circulation, which has provided significant motivation for utilizing proximal fluids as a primary sample source for protein biomarker discovery. The present review highlights recent advances in proximal fluid proteomics, including the various protocols utilized to harvest proximal fluids along with detailing the results from mass spectrometry- and antibody-based analyses.
Collapse
Affiliation(s)
- Pang-ning Teng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|