1
|
Patel A, Dewani D, Jaiswal A, Reddy LS, Yadav P, Sethi N. Unveiling the Role of Pregnancy-Associated Plasma Protein A (PAPP-A) in Pregnancy-Associated Breast Cancer: A Comprehensive Review. Cureus 2024; 16:e56269. [PMID: 38623138 PMCID: PMC11017795 DOI: 10.7759/cureus.56269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/16/2024] [Indexed: 04/17/2024] Open
Abstract
Pregnancy-associated breast cancer (PABC) presents unique challenges due to its occurrence during or shortly after pregnancy. Pregnancy-associated plasma protein A (PAPP-A) has emerged as a potential biomarker and regulator in PABC. This comprehensive review examines the role of PAPP-A in PABC, highlighting its involvement in tissue remodeling and cancer progression. Molecular mechanisms linking PAPP-A to breast cancer, including signaling pathways and interactions with other molecules, are explored. The review also discusses the diagnostic and therapeutic implications of PAPP-A dysregulation in PABC, emphasizing the need for further research to elucidate underlying mechanisms and develop targeted therapies. Collaborative efforts among researchers, clinicians, and industry stakeholders are essential for translating findings into clinically relevant interventions to improve outcomes for PABC patients.
Collapse
Affiliation(s)
- Archan Patel
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Deepika Dewani
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arpita Jaiswal
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Lucky Srivani Reddy
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Pallavi Yadav
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Neha Sethi
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
2
|
Poddar A, Ahmady F, Rao SR, Sharma R, Kannourakis G, Prithviraj P, Jayachandran A. The role of pregnancy associated plasma protein-A in triple negative breast cancer: a promising target for achieving clinical benefits. J Biomed Sci 2024; 31:23. [PMID: 38395880 PMCID: PMC10885503 DOI: 10.1186/s12929-024-01012-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Pregnancy associated plasma protein-A (PAPP-A) plays an integral role in breast cancer (BC), especially triple negative breast cancer (TNBC). This subtype accounts for the most aggressive BC, possesses high tumor heterogeneity, is least responsive to standard treatments and has the poorest clinical outcomes. There is a critical need to address the lack of effective targeted therapeutic options available. PAPP-A is a protein that is highly elevated during pregnancy. Frequently, higher PAPP-A expression is detected in tumors than in healthy tissues. The increase in expression coincides with increased rates of aggressive cancers. In BC, PAPP-A has been demonstrated to play a role in tumor initiation, progression, metastasis including epithelial-mesenchymal transition (EMT), as well as acting as a biomarker for predicting patient outcomes. In this review, we present the role of PAPP-A, with specific focus on TNBC. The structure and function of PAPP-A, belonging to the pappalysin subfamily, and its proteolytic activity are assessed. We highlight the link of BC and PAPP-A with respect to the IGFBP/IGF axis, EMT, the window of susceptibility and the impact of pregnancy. Importantly, the relevance of PAPP-A as a TNBC clinical marker is reviewed and its influence on immune-related pathways are explored. The relationship and mechanisms involving PAPP-A reveal the potential for more treatment options that can lead to successful immunotherapeutic targets and the ability to assist with better predicting clinical outcomes in TNBC.
Collapse
Affiliation(s)
- Arpita Poddar
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
- RMIT University, Victoria, Australia
| | - Farah Ahmady
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Sushma R Rao
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Revati Sharma
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Prashanth Prithviraj
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Victoria, Australia.
- Federation University, Victoria, Australia.
| |
Collapse
|
3
|
Wang D, Peng H, Hu Y, Piao X, Gao D, Sha Y. Distinctive gene expression patterns in pregnancy-associated breast cancer. Front Genet 2022; 13:850195. [PMID: 36035177 PMCID: PMC9399642 DOI: 10.3389/fgene.2022.850195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Pregnancy-associated breast cancer (PABC) is diagnosed during pregnancy or within 1 year postpartum, but the unique aspects of its etiology and pathogenesis have not been fully elucidated. This study aimed to ascertain the molecular mechanisms of PABC to facilitate diagnosis and therapeutic development. The Limma package was used to characterize the differentially expressed genes in PABC as compared to non-pregnancy-associated breast cancer (NPABC) and normal breast tissue. A total of 871 dysregulated genes were identified in the PABC versus NPABC groups and 917 in the PABC versus normal groups, with notable differences in the expression of MAGE and CXCL family genes. The dysregulated genes between the PABC and normal groups were mainly associated with signal transduction and immune response, while Kyoto Encyclopedia of Genes and Genomes analysis revealed that the dysregulated genes were enriched in immune-related pathways, including the major histocompatibility complex (MHC) class II protein complex, the type I interferon signaling pathway, regulation of α-β T-cell proliferation, and the T-cell apoptotic process. Through protein-protein interaction network construction, CD44 and BRCA1 were identified as prominent hub genes with differential expression in PABC versus NPABC. Furthermore, a cluster with eleven hub genes was identified in PABC versus normal adjacent tissues, of which the expression of EGFR, IGF1, PTGS2, FGF1, CAV1, and PLCB1 were verified to be differentially expressed in an independent cohort of PABC patients. Notably, IGF1, PTGS2, and FGF1 were demonstrated to be significantly related to patient prognosis. Our study reveals a distinctive gene expression pattern in PABC and suggests that IGF1, PTGS2, and FGF1 might serve as biomarkers for diagnosis and prognosis of PABC.
Collapse
Affiliation(s)
- Dan Wang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Huiyu Peng
- The Key Laboratory of BioMedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Suzhou, China
| | - Yuyao Hu
- School of Life Science, Xuzhou Medical University, Xuzhou, China
| | - Xue Piao
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Research Center for Neurobiology of Xuzhou Medical University, Xuzhou, China
| | - Yan Sha
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yan Sha,
| |
Collapse
|
4
|
Gómez-Cuadrado L, Bullock E, Mabruk Z, Zhao H, Souleimanova M, Noer PR, Turnbull AK, Oxvig C, Bertos N, Byron A, Dixon JM, Park M, Haider S, Natrajan R, Sims AH, Brunton VG. Characterisation of the Stromal Microenvironment in Lobular Breast Cancer. Cancers (Basel) 2022; 14:904. [PMID: 35205651 PMCID: PMC8870100 DOI: 10.3390/cancers14040904] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 02/06/2022] [Indexed: 11/16/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is the second most common histological subtype of breast cancer, and it exhibits a number of clinico-pathological characteristics distinct from the more common invasive ductal carcinoma (IDC). We set out to identify alterations in the tumor microenvironment (TME) of ILC. We used laser-capture microdissection to separate tumor epithelium from stroma in 23 ER+ ILC primary tumors. Gene expression analysis identified 45 genes involved in regulation of the extracellular matrix (ECM) that were enriched in the non-immune stroma of ILC, but not in non-immune stroma from ER+ IDC or normal breast. Of these, 10 were expressed in cancer-associated fibroblasts (CAFs) and were increased in ILC compared to IDC in bulk gene expression datasets, with PAPPA and TIMP2 being associated with better survival in ILC but not IDC. PAPPA, a gene involved in IGF-1 signaling, was the most enriched in the stroma compared to the tumor epithelial compartment in ILC. Analysis of PAPPA- and IGF1-associated genes identified a paracrine signaling pathway, and active PAPP-A was shown to be secreted from primary CAFs. This is the first study to demonstrate molecular differences in the TME between ILC and IDC identifying differences in matrix organization and growth factor signaling pathways.
Collapse
Affiliation(s)
- Laura Gómez-Cuadrado
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Esme Bullock
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Zeanap Mabruk
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Hong Zhao
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Margarita Souleimanova
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark; (P.R.N.); (C.O.)
| | - Arran K. Turnbull
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark; (P.R.N.); (C.O.)
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - J. Michael Dixon
- Edinburgh Breast Unit, University of Edinburgh, Edinburgh EH4 2XU, UK;
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.H.); (R.N.)
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.H.); (R.N.)
| | - Andrew H. Sims
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Valerie G. Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| |
Collapse
|
5
|
Marzocchi C, Capezzone M, Sagnella A, Cartocci A, Caroli Costantini M, Brindisi L, Mancini V, Cantara S, Castagna MG. Pregnancy-associated plasma protein A mRNA expression as a marker for differentiated thyroid cancer: results from a "surgical" and a "cytological" series. J Endocrinol Invest 2022; 45:369-378. [PMID: 34350538 PMCID: PMC8783868 DOI: 10.1007/s40618-021-01655-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/29/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE Pregnancy-associated plasma protein A (PAPPA) is a metalloproteinase initially described for its role during pregnancy. PAPPA regulates IGF ligands 1 (IGF1) bioavailability through the degradation of IGF-binding protein 4 (IGFBP4). After the cleavage of IGFBP4, free IGF1 is able to bind IGF1 receptors (IGF1R) triggering the downstream signaling. Recently, PAPPA expression has been linked with development of several cancers. No data have been published on thyroid cancer, yet. METHODS We evaluated PAPPA, insulin-like growth factor (IGF1), IGF1 receptors (IGF1R) and IGF-binding protein 4 (IGFBP4) mRNA expression levels in a "Surgical series" of 94 thyroid nodules (64 cancers, 16 follicular adenomas and 14 hyperplastic nodules) and in a "Cytological series" of 80 nodules from 74 patients underwent to fine-needle aspiration cytology (FNAC). In tissues, PAPPA was also evaluated by western blot. RESULTS We found that PAPPA expression was increased in thyroid cancer specimen at mRNA and protein levels and that, adenomas and hyperplastic nodules had an expression similar to normal tissues. When applied on thyroid cytologies, PAPPA expression was able to discriminate benign from malignant nodules contributing to pre-surgical classification of the nodules. We calculated a cut-off with a good specificity (91%) which reached 100% when combined with molecular biology. CONCLUSION These results show that PAPPA could represent a promising diagnostic marker for differentiated thyroid cancer.
Collapse
Affiliation(s)
- C Marzocchi
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - M Capezzone
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - A Sagnella
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - A Cartocci
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - M Caroli Costantini
- Department of Oncology and Pathological Anatomy, Azienda Ospedaliera, Universitario Senese, Siena, Italy
| | - L Brindisi
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - V Mancini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - S Cantara
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy.
| | - M G Castagna
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| |
Collapse
|
6
|
Ma C, Wang Y, Wilson KM, Mucci LA, Stampfer MJ, Pollak M, Penney KL. Circulating Insulin-Like Growth Factor 1-Related Biomarkers and Risk of Lethal Prostate Cancer. JNCI Cancer Spectr 2022; 6:pkab091. [PMID: 35047751 PMCID: PMC8763370 DOI: 10.1093/jncics/pkab091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/26/2021] [Accepted: 10/21/2021] [Indexed: 11/22/2022] Open
Abstract
Background Experimental and epidemiologic evidence supports the role of circulating insulin-like growth factor-1 (IGF-1) levels with the risk of prostate cancer. Most circulating IGF-1 is bound to specific binding proteins, and only about 5% circulates in a free form. We explored the relation of free IGF-1 and other components of the IGF system with lethal prostate cancer. Methods Using prospectively collected samples, we undertook a nested case-only analysis among 434 men with lethal prostate cancer and 524 men with indolent, nonlethal prostate cancer in the Physicians’ Health Study and the Health Professionals Follow-up Study. Prediagnostic plasma samples were assayed for free IGF-1 and total IGF-1, acid labile subunit, pregnancy-associated plasma protein A (PAPP-A), and intact and total IGF binding protein 4. We estimated odds ratios (ORs) and corresponding 95% confidence intervals (CIs) for the associations between IGF-1–related biomarkers and lethal prostate cancer using unconditional logistic regression models adjusted for age, height, and body mass index. Results Men in the highest quartile of PAPP-A levels had 42% higher odds of lethal prostate cancer (pooled adjusted OR = 1.42, 95% CI = 1.04 to 1.92) compared with men in the lowest 3 quartiles. There were no statistically significant differences in the other plasma analytes. The positive association between PAPP-A and lethal prostate cancer was present among men with intact PTEN but not among those with tumor PTEN loss (2-sided Pinteraction = .001). Conclusions Our study provides suggestive evidence that among men who later develop prostate cancer, higher plasma PAPP-A levels measured prior to diagnosis are associated with increased risk of lethal compared with indolent disease.
Collapse
Affiliation(s)
- Chaoran Ma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Correspondence to: Chaoran Ma, MD, PhD, Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA 02115, USA (e-mail: )
| | - Ye Wang
- Oncology Department, McGill University and Segal Cancer Centre, Jewish General Hospital, Montreal, QC, Canada
| | - Kathryn M Wilson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael Pollak
- Oncology Department, McGill University and Segal Cancer Centre, Jewish General Hospital, Montreal, QC, Canada
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
7
|
Zhang J, Zhang Y, Li L, Nian Y, Chen Y, Shen R, Ma X. Pregnancy-associated plasma protein-A (PAPPA) promotes breast cancer progression. Bioengineered 2022; 13:291-307. [PMID: 34974815 PMCID: PMC8805841 DOI: 10.1080/21655979.2021.2000724] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Breast cancer is the most common malignancy in females and poses a significant health threat to women. Pregnancy-associated plasma protein-A (PAPPA) is highly expressed in pregnancy-associated breast cancer (PABC) tissues. In this study, we investigated the functional role of PAPPA in regulating the malignant phenotype of breast cancer. We first examined the expression level of PAPPA in PABC tissue and breast cancer cell lines using quantitative real-time polymerase-chain reaction (qRT-PCR) and western blot. Next, the functional role of PAPPA in breast cancer cells was validated by overexpression and knockdown experiments. Cell counting kit-8 (CCK-8) proliferation assay, 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay, wound healing and transwell invasion assay were used to examine cell proliferation, migration, and invasion ability. We further identified the microRNA target regulating PAPPA and studied its functional role. Finally, we examined the impact of PAPPA on the tumorigenesis and metastasis of breast cancer in mice model. Our study revealed that PAPPA was upregulated in PABC tissues and breast cancer cells. Overexpression of PAPPA promoted cell proliferation, motility, invasion, and epithelial-mesenchymal transition (EMT). We further identified miR-497-5p as a negative regulator of PAPPA, which suppressed cell proliferation, migration, invasion, and EMT in breast cancer cells. We also validated the oncogenic role of PAPPA in the mouse xenograft model. Collectively, our study suggests that PAPPA is an oncogenic protein highly expressed in PABC tissues and promotes breast cancer progression, which could serve as a novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Clinical Laboratory, The Fifth Hospital of Wuhan, Wuhan, China
| | - Yuan Zhang
- Department of Ultrasound Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lanjiang Li
- Department of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yinghua Nian
- Department of Ultrasound Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Kunming Medical University, Kunming, China
| | - Ruoxia Shen
- Department of Ultrasound Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaoyan Ma
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Lu Y, Li S, Wang T, Liao X, Mao L, Li Z. PAPP-A functions as a tumor suppressor and is downregulated in renal cell carcinoma. FEBS Open Bio 2021; 11:1593-1606. [PMID: 33788403 PMCID: PMC8167875 DOI: 10.1002/2211-5463.13156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 01/17/2023] Open
Abstract
Pregnancy‐associated plasma protein A (PAPP‐A) is a proteolytic enzyme produced by the placenta. The expression and role of PAPP‐A in renal cell carcinoma (RCC) remain elusive. The aim of this study was to investigate the role and the molecular mechanisms of PAPP‐A in RCC. Initially, we evaluated the expression of PAPP‐A in samples from patients with RCC and cell lines by quantitative PCR, western blot and immunohistochemical staining, and examined the role of PAPP‐A in RCC cells by cell viability, colony formation and Transwell assays. Next, we investigated the molecular mechanisms regulating the tumor suppressor function of PAPP‐A. Our results demonstrated that PAPP‐A is expressed at low levels in RCC tissues and cells. Clinical data analysis revealed a significant correlation between PAPP‐A expression and RCC‐related death (P < 0.0115). Overexpression of PAPP‐A inhibited viability, proliferation, migration and invasion of RCC cells. Furthermore, PAPP‐A overexpression significantly increased phosphorylation of c‐Jun N‐terminal kinase and decreased the expression of cyclin D1, phosphorylated glycogen synthase kinase‐3β and β‐catenin. This study is the first to report that downregulation of PAPP‐A is associated with poor prognosis in patients with RCC. In conclusion, PAPP‐A may serve as a novel prognostic marker and potentially as a therapeutic target in patients with RCC.
Collapse
Affiliation(s)
- Yanxin Lu
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Zhuhai Campus of Zunyi Medical University, China.,Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Shi Li
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China
| | - Tongyu Wang
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China
| | - Ximian Liao
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China
| | - Longyi Mao
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China
| | - Zesong Li
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, China
| |
Collapse
|
9
|
Prithviraj P, Anaka M, Thompson EW, Sharma R, Walkiewicz M, Tutuka CSA, Behren A, Kannourakis G, Jayachandran A. Aberrant pregnancy-associated plasma protein-A expression in breast cancers prognosticates clinical outcomes. Sci Rep 2020; 10:13779. [PMID: 32792532 PMCID: PMC7426935 DOI: 10.1038/s41598-020-70774-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/03/2020] [Indexed: 01/15/2023] Open
Abstract
Elevated levels of pregnancy-associated plasma protein-A (PAPP-A) have been implicated in the pathogenesis of various malignancies, including breast cancers. Breast cancer is one of the most frequent carcinomas and is the second most common cancer type detected in women of child-bearing age. Throughout pregnancy PAPP-A is produced and secreted by the placental syncytiotrophoblast cells; co-incidentally pregnancy-associated breast cancers often have an aggressive clinical course. The components of the PAPP-A/IGF axis was assessed in a panel of breast cancer cell lines. Using neutralising antibodies the impact of PAPP-A/IGF axis on cell motility was evaluated. PAPP-A was expressed in four of the twelve breast cancer cell lines tested. Blocking PAPP-A and IGFBP4 with neutralising antibodies significantly decreased motiliy of MDA-MB-231 cells. Upregulation of PAPP-A expression in breast tumours resulted in a trend towards worse overall survival. Notably, PAPP-A expression also positively correlated with epithelial-to-mesenchymal transition markers. In conclusion, these results indicate that PAPP-A plays an important role in breast cancer progression and it may be a promising therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Prashanth Prithviraj
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia.,Fiona Elsey Cancer Research Institute, Ballarat Technology Park- Central Suite 23, 106-110 Lydiard St Sth, Ballarat, VIC, 3350, Australia.,Federation University Australia, Ballarat, VIC, Australia
| | - Matthew Anaka
- Department of Medicine, University of Alberta, Alberta, Canada
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat Technology Park- Central Suite 23, 106-110 Lydiard St Sth, Ballarat, VIC, 3350, Australia.,Federation University Australia, Ballarat, VIC, Australia
| | - Marzena Walkiewicz
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Candani S A Tutuka
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Victoria, Australia
| | - Andreas Behren
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Victoria, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat Technology Park- Central Suite 23, 106-110 Lydiard St Sth, Ballarat, VIC, 3350, Australia.,Federation University Australia, Ballarat, VIC, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Ballarat Technology Park- Central Suite 23, 106-110 Lydiard St Sth, Ballarat, VIC, 3350, Australia. .,Gallipoli Medical Research Institute and The University of Queensland, Brisbane, Australia.
| |
Collapse
|
10
|
Kashyap S, Hein KZ, Chini CC, Lika J, Warner GM, Bale LK, Torres VE, Harris PC, Oxvig C, Conover CA, Chini EN. Metalloproteinase PAPP-A regulation of IGF-1 contributes to polycystic kidney disease pathogenesis. JCI Insight 2020; 5:135700. [PMID: 31990681 DOI: 10.1172/jci.insight.135700] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/22/2020] [Indexed: 02/06/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic cause of end-stage renal disease (ESRD). The treatment options for ADPKD are limited. We observed an upregulation in several IGF-1 pathway genes in the kidney of Pkd1RC/RC mice, a model of ADPKD. Pregnancy-associated plasma protein A (PAPP-A), a metalloproteinase that cleaves inhibitory IGF binding proteins (IGFBPs), increasing the local bioactivity of IGF-1, was highly induced in the kidney of ADPKD mice. PAPP-A levels were high in cystic fluid and kidneys of humans with ADPKD. Our studies further showed that PAPP-A transcription in ADPKD was mainly regulated through the cAMP/CREB/CBP/p300 pathway. Pappa deficiency effectively inhibited the development of cysts in the Pkd1RC/RC mice. The role of PAPP-A in cystic disease appears to be regulation of the IGF-1 pathway and cellular proliferation in the kidney. Finally, preclinical studies demonstrated that treatment with a monoclonal antibody that blocks the proteolytic activity of PAPP-A against IGFBP4 ameliorated ADPKD cystic disease in vivo in Pkd1RC/RC mice and ex vivo in embryonic kidneys. These data indicated that the PAPP-A/IGF-1 pathway plays an important role in the growth and expansion of cysts in ADPKD. Our findings introduce a therapeutic strategy for ADPKD that involves the inhibition of PAPP-A.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| | - Kyaw Zaw Hein
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| | - Claudia Cs Chini
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| | - Jorgo Lika
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| | - Gina M Warner
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| | - Laurie K Bale
- Division of Endocrinology and Metabolism, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension and Robert M. and Billie Kelley Pirnie Translational PKD Center, Rochester, Minnesota, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension and Robert M. and Billie Kelley Pirnie Translational PKD Center, Rochester, Minnesota, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Cheryl A Conover
- Division of Endocrinology and Metabolism, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| | - Eduardo N Chini
- Department of Anesthesiology and Robert and Arlene Kogod Center on Aging
| |
Collapse
|
11
|
Bøtkjær JA, Pors SE, Petersen TS, Kristensen SG, Jeppesen JV, Oxvig C, Andersen CY. Transcription profile of the insulin-like growth factor signaling pathway during human ovarian follicular development. J Assist Reprod Genet 2019; 36:889-903. [PMID: 30877600 DOI: 10.1007/s10815-019-01432-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/27/2019] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The IGF signaling cascade exerts important regulatory functions in human ovarian folliculogenesis. The scope of this study was to evaluate the transcription profile of insulin-like growth factor (IGF) genes during human ovarian follicle development and to analyze follicle fluid levels of key IGF proteins. METHODS Gene expression profiling was performed with microarray gene analysis. The analysis was assessed from ovarian follicles and granulosa cells (GCs) obtained from isolated stage-specific human ovarian follicles, including preantral follicles, small antral follicles, and preovulatory follicles. Numerous genes involved in the IGF signaling pathway was evaluated and key genes were validated by qPCR from GCs. Protein levels of various IGF components of human follicular fluid (FF) were measured by ELISA and time-resolved immunofluorometric assays (TRIFMA). RESULTS The gene expression levels of PAPPA, IGF2, IGF receptors and intracellular IGF-activated genes increased with increasing follicle size. This was especially prominent in the late preovulatory stage where IGF2 expression peaked. Protein levels of intact IGF binding protein-4 decreased significantly in FF from large preovulatory follicles compared with small antral follicles concomitant with higher protein levels of PAPP-A. The IGF modulators IGF-2 receptor, IGFBPs, stanniocalcins, and IGF-2 mRNA binding proteins were all observed to be expressed in the different follicle stages. CONCLUSIONS This study confirms and highlights the importance of PAPP-A regulating bioactive IGF levels throughout folliculogenesis and especially for the high rate of granulosa cell proliferation and expression of key ovarian hormones important in the last part of the follicular phase of the menstrual cycle.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark.
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark
| | - Tonny Studsgaard Petersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark
| | - Janni Vikkelsø Jeppesen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000, Aarhus, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, DK-2100, Copenhagen, Denmark
| |
Collapse
|
12
|
Hjortebjerg R. IGFBP-4 and PAPP-A in normal physiology and disease. Growth Horm IGF Res 2018; 41:7-22. [PMID: 29864720 DOI: 10.1016/j.ghir.2018.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Insulin-like growth factor (IGF) binding protein-4 (IGFBP-4) is a modulator of the IGF system, exerting both inhibitory and stimulatory effects on IGF-induced cellular growth. IGFBP-4 is the principal substrate for the enzyme pregnancy-associated plasma protein-A (PAPP-A). Through IGF-dependent cleavage of IGFBP-4 in the vicinity of the IGF receptor, PAPP-A is able to increase IGF bioavailability and stimulate IGF-mediated growth. Recently, the stanniocalcins (STCs) were identified as novel inhibitors of PAPP-A proteolytic activity, hereby adding additional members to the seemingly endless list of proteins belonging to the IGF family. Our understanding of these proteins has advanced throughout recent years, and there is evidence to suggest that the role of IGFBP-4 and PAPP-A in defining the relationship between total IGF and IGF bioactivity can be linked to a number of pathological conditions. This review provides an overview of the experimental and clinical findings on the IGFBP-4/PAPP-A/STC axis as a regulator of IGF activity and examines the conundrum surrounding extrapolation of circulating concentrations to tissue action of these proteins. The primary focus will be on the biological significance of IGFBP-4 and PAPP-A in normal physiology and in pathophysiology with emphasis on metabolic disorders, cardiovascular diseases, and cancer. Finally, the review assesses current new trajectories of IGFBP-4 and PAPP-A research.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| |
Collapse
|
13
|
Abstract
The zinc metalloproteinase, PAPP-A, enhances local insulin-like growth factor (IGF) action through cleavage of inhibitory IGF-binding proteins, thereby increasing IGF available for IGF receptor-mediated cell proliferation, migration and survival. In many tumors, enhanced IGF receptor signaling is associated with tumor growth, invasion and metastasis. We will first discuss PAPP-A structure and function, and post-translational inhibitors of PAPP-A expression or proteolytic activity. We will then review the evidence supporting an important role for PAPP-A in many cancers, including breast, ovarian and lung cancer, and Ewing sarcoma.
Collapse
Affiliation(s)
- Cheryl A Conover
- From the Division of Endocrinology Mayo ClinicRochester, Minnesota, USA
| | - Claus Oxvig
- Department of Molecular Biology and GeneticsAarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Saavedra-García P, Nichols K, Mahmud Z, Fan LYN, Lam EWF. Unravelling the role of fatty acid metabolism in cancer through the FOXO3-FOXM1 axis. Mol Cell Endocrinol 2018; 462:82-92. [PMID: 28087388 DOI: 10.1016/j.mce.2017.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/06/2016] [Accepted: 01/09/2017] [Indexed: 02/07/2023]
Abstract
Obesity and cachexia represent divergent states of nutritional and metabolic imbalance but both are intimately linked to cancer. There is an extensive overlap in their signalling pathways and molecular components involved such as fatty acids (FAs), which likely play a crucial role in cancer. Forkhead box (FOX) proteins are responsible of a wide range of transcriptional programmes during normal development, and the FOXO3-FOXM1 axis is associated with cancer initiation, progression and drug resistance. Free fatty acids (FFAs), FA synthesis and β-oxidation are associated with cancer development and progression. Meanwhile, insulin and some adipokines, that are up-regulated by FAs, are also involved in cancer development and poor prognosis. In this review, we discuss the role of FA metabolism in cancer and how FA metabolism integrates with the FOXO3-FOXM1 axis. These new insights may provide leads to better cancer diagnostics as well as strategies for tackling cancer development, progression and drug resistance.
Collapse
Affiliation(s)
- Paula Saavedra-García
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Katie Nichols
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Zimam Mahmud
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Lavender Yuen-Nam Fan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| |
Collapse
|
15
|
de Cristofaro T, Di Palma T, Soriano AA, Monticelli A, Affinito O, Cocozza S, Zannini M. Candidate genes and pathways downstream of PAX8 involved in ovarian high-grade serous carcinoma. Oncotarget 2018; 7:41929-41947. [PMID: 27259239 PMCID: PMC5173106 DOI: 10.18632/oncotarget.9740] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/16/2016] [Indexed: 12/26/2022] Open
Abstract
Understanding the biology and molecular pathogenesis of ovarian epithelial cancer (EOC) is key to developing improved diagnostic and prognostic indicators and effective therapies. Although research has traditionally focused on the hypothesis that high-grade serous carcinoma (HGSC) arises from the ovarian surface epithelium (OSE), recent studies suggest that additional sites of origin exist and a substantial proportion of cases may arise from precursor lesions located in the Fallopian tubal epithelium (FTE). In FTE cells, the transcription factor PAX8 is a marker of the secretory cell lineage and its expression is retained in 96% of EOC. We have recently reported that PAX8 is involved in the tumorigenic phenotype of ovarian cancer cells. In this study, to uncover genes and pathways downstream of PAX8 involved in ovarian carcinoma we have determined the molecular profiles of ovarian cancer cells and in parallel of Fallopian tube epithelial cells by means of a silencing approach followed by an RNA-seq analysis. Interestingly, we highlighted the involvement of pathways like WNT signaling, epithelial-mesenchymal transition, p53 and apoptosis. We believe that our analysis has led to the identification of candidate genes and pathways regulated by PAX8 that could be additional targets for the therapy of ovarian carcinoma.
Collapse
Affiliation(s)
- Tiziana de Cristofaro
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Tina Di Palma
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Amata Amy Soriano
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Antonella Monticelli
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Ornella Affinito
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Sergio Cocozza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Mariastella Zannini
- IEOS, Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| |
Collapse
|
16
|
Mak CSL, Yung MMH, Hui LMN, Leung LL, Liang R, Chen K, Liu SS, Qin Y, Leung THY, Lee KF, Chan KKL, Ngan HYS, Chan DW. MicroRNA-141 enhances anoikis resistance in metastatic progression of ovarian cancer through targeting KLF12/Sp1/survivin axis. Mol Cancer 2017; 16:11. [PMID: 28095864 PMCID: PMC5240442 DOI: 10.1186/s12943-017-0582-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer metastasis is determined by the formation of the metastatic niche and the ability of cancer cells to adapt to microenvironmental stresses. Anoikis resistance is a fundamental feature of metastatic cancer cell survival during metastatic cancer progression. However, the mechanisms underlying anoikis resistance in ovarian cancer are still unclear. METHODS Expressions of miRNA-141 and its downstream targets were evaluated by qPCR, Western blotting, Immunohistochemical (IHC) and in situ hybridization (ISH) assays. The luciferase assays were used to prove KLF12 as the downstream target of miR-141. The cDNA microarray and apoptotic protein arrays were used to identify the targets of miR-141 and KLF12. The competition of KLF12 and Sp1 on survivin promoter was examined by ChIP assay. IHC analysis on ovarian cancer tissue array was used to evaluate the expressions of KLF12 and miR-141 and to show the clinical relevance. The functional studies were performed by in vitro and in vivo tumorigenic assays. RESULTS Enforced expression of miR-141 promotes, while knockdown of miR-141 expression inhibits, cell proliferation, anchorage-independent capacity, anoikis resistance, tumor growth and peritoneal metastases of ovarian cancer cells. Bioinformatics and functional analysis identified that Kruppel-related zinc finger protein AP-2rep (KLF12) is directly targeted by miR-141. Consistent with this finding, knockdown of KLF12 phenocopied the effects of miR-141 overexpression in ovarian cancer cells. In contrast, restoration of KLF12 in miR-141-expressing cells significantly attenuated anoikis resistance in ovarian cancer cells via interfering with Sp1-mediated survivin transcription, which inhibits the intrinsic apoptotic pathway and is crucial for ovarian cancer cell survival, anoikis resistance and peritoneal metastases. Immunohistochemical (IHC) and in situ hybridization (ISH) assays confirmed that miRNA-141 expression is inversely correlated with KLF12 expression and significantly associated with advanced ovarian cancers accompanied with distal metastases, underscoring the clinical relevance of our findings. CONCLUSIONS Our data identify a novel signaling axis of miR-141/KLF12/Sp1/survivin in enhancing anoikis resistance and likely serves as a potential therapeutic target for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Celia S L Mak
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Mingo M H Yung
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Lynn M N Hui
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Leanne L Leung
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Rui Liang
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Kangmei Chen
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Stephanie S Liu
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Yiming Qin
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Thomas H Y Leung
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Karen K L Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China
| | - David W Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
17
|
Kirschner A, Thiede M, Grünewald TGP, Alba Rubio R, Richter GHS, Kirchner T, Busch DH, Burdach S, Thiel U. Pappalysin-1 T cell receptor transgenic allo-restricted T cells kill Ewing sarcoma in vitro and in vivo. Oncoimmunology 2017; 6:e1273301. [PMID: 28344885 PMCID: PMC5353903 DOI: 10.1080/2162402x.2016.1273301] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 01/06/2023] Open
Abstract
Pregnancy-associated plasma protein-A (PAPPA), also known as pappalysin, is a member of the insulin-like growth factor (IGF) family. PAPPA acts as a protease, cleaving IGF inhibitors, i.e., IGF binding proteins (IGFBPs), thereby setting free IGFs. The insulin/IGF-axis is involved in cancer in general and in Ewing sarcoma (ES) in particular. ES is a highly malignant bone tumor characterized by early metastatic spread. PAPPA is associated with various cancers. It is overexpressed and required for proliferation in ES. PAPPA also stimulates normal bone growth. We isolated HLA-A*02:01+/peptide-restricted T cells from A*02:01− healthy donors directed against PAPPA, generated by priming with A*02:01+ PAPPA peptide loaded dendritic cells. After TCR identification, retrovirally TCR transduced CD8+ T cells were assessed for their in vitro specificity and in vivo efficacy in human ES bearing Rag2−/−γc−/− mice. Engraftment in mice and tumor infiltration of TCR transgenic T cells in the mice was evaluated. The TCR transgenic T cell clone PAPPA-2G6 demonstrated specific reactivity toward HLA-A*02:01+/PAPPA+ ES cell lines. We furthermore detected circulating TCR transgenic T cells in the blood in Rag2−/−γc−/− mice and in vivo engraftment in bone marrow. Tumor growth in mice with xenografted ES was significantly reduced after treatment with PAPPA-2G6 TCR transgenic T cells in contrast to controls. Tumors of treated mice revealed tumor-infiltrating PAPPA-2G6 TCR transgenic T cells. In summary, we demonstrate that PAPPA is a first-rate target for TCR-based immunotherapy of ES.
Collapse
Affiliation(s)
- Andreas Kirschner
- Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children's Cancer Research Center, Klinikum rechts der Isar, Technische Universität München , München, Germany
| | - Melanie Thiede
- Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children's Cancer Research Center, Klinikum rechts der Isar, Technische Universität München , München, Germany
| | - Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology of the LMU Munich, München, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebeca Alba Rubio
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology of the LMU Munich , München, Germany
| | - Günther H S Richter
- Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children's Cancer Research Center, Klinikum rechts der Isar, Technische Universität München, München, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Kirchner
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology of the LMU Munich, München, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Comprehensive Cancer Center (CCC) Munich, München, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , München, Germany
| | - Stefan Burdach
- Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children's Cancer Research Center, Klinikum rechts der Isar, Technische Universität München, München, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Comprehensive Cancer Center (CCC) Munich, München, Germany
| | - Uwe Thiel
- Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children's Cancer Research Center, Klinikum rechts der Isar, Technische Universität München , München, Germany
| |
Collapse
|
18
|
Kazarian A, Blyuss O, Metodieva G, Gentry-Maharaj A, Ryan A, Kiseleva EM, Prytomanova OM, Jacobs IJ, Widschwendter M, Menon U, Timms JF. Testing breast cancer serum biomarkers for early detection and prognosis in pre-diagnosis samples. Br J Cancer 2017; 116:501-508. [PMID: 28081538 PMCID: PMC5318971 DOI: 10.1038/bjc.2016.433] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/14/2016] [Accepted: 12/08/2016] [Indexed: 02/08/2023] Open
Abstract
Background: Breast cancer is a leading cause of morbidity and mortality worldwide. Although mammography screening is available, there is an ongoing interest in improved early detection and prognosis. Herein, we have analysed a combination of serological biomarkers in a case–control cohort of sera taken before diagnosis. Methods: This nested case–control study within the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) used serum samples from 239 women who subsequently developed breast cancer and 239 matched cancer-free controls. Sera were screened by ELISA for 9 candidate markers. Univariate and multivariate analyses were performed to examine associations with clinico-pathological features and between case controls in different time groups before diagnosis. Results: Significant associations with clinico-pathological features related to prognosis were found for several candidates (CA15-3, HSP90A and PAI-1). However, there were no consistent differences between cases and controls for any candidate in the lead up to diagnosis. Whilst combination models outperformed single markers, there was no increase in performance towards diagnosis. Conclusions: This study using unique pre-diagnosis samples shows that CA15-3, HSP90A and PAI-1 have potential as early prognostic markers and warrant further investigation. However, none of the candidates or combinations would be useful for screening.
Collapse
Affiliation(s)
- Anna Kazarian
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - Oleg Blyuss
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - Gergana Metodieva
- Department of Biological Sciences, University of Essex, Colchester, Essex CO4 3SQ, UK
| | - Aleksandra Gentry-Maharaj
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - Andy Ryan
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - Elena M Kiseleva
- Oles Honchar Dnipropetrovsk National University, Dnipropetrovsk, Ukraine
| | - Olga M Prytomanova
- Oles Honchar Dnipropetrovsk National University, Dnipropetrovsk, Ukraine
| | - Ian J Jacobs
- University of New South Wales, Sydney, NSW 2052, Australia
| | - Martin Widschwendter
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - Usha Menon
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| | - John F Timms
- Department of Women's Cancer, Institute for Women's Health, University College London, London, W1T 7DN, UK
| |
Collapse
|
19
|
Bøtkjær JA, Borgbo T, Kløverpris S, Noer PR, Oxvig C, Andersen CY. Effect of pregnancy-associated plasma protein-A (PAPP-A) single-nucleotide polymorphisms on the level and activity of PAPP-A and the hormone profile in fluid from normal human small antral follicles. Fertil Steril 2016; 106:1778-1786.e8. [PMID: 27793387 DOI: 10.1016/j.fertnstert.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/10/2016] [Accepted: 09/07/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To reveal a possible relationship between two single nucleotide polymorphisms (SNPs) in PAPP-A-1224 (rs7020782) and 327 (rs12375498)-and the level and activity of PAPP-A in follicular fluid (FF) of human small antral follicles, and to analyze the intrafollicular hormone levels. DESIGN Laboratory investigation. SETTING University hospital. PATIENT(S) Fifty volunteer women who contributed a total of 210 samples of FF from normal small antral follicles. INTERVENTION(S) Genotyping and measurement of antigen levels of steroids, PAPP-A, stanniocalcin-2 (STC2), and antimüllerian hormone (AMH) plus activity of PAPP-A toward insulin-like growth factor binding protein 4 (IGFBP-4). MAIN OUTCOME MEASURE(S) Measurement of PAPP-A levels and hormones with enzyme-linked immunosorbent assay (ELISA) and PAPP-A activity toward radiolabeled IGFBP-4. RESULT(S) Women homozygous for the minor C allele of the 1224 SNP showed a statistically significantly lower level of PAPP-A protein and activity in FF compared with women carrying the major A allele. These women also displayed nonsignificant reduced levels of estradiol and increased levels of AMH and androgen. A statistically significant correlation between FF levels of PAPP-A activity and the molar ratio of PAPP-A/STC2 was obtained. The 327 SNP did not show statistically significant associations. CONCLUSION(S) This study presents a statistically significant effect of the 1224 SNP on the level and activity of PAPP-A in human follicles, suggesting that the FF level of bioactive insulin-like growth factor depends on the genotype. We observed STC2 to be an important regulator of PAPP-A in human FF. The 1224 SNP has previously been associated with recurrent pregnancy loss, so further evaluation of an underlying mechanism including aberrant control of insulin-like growth factor activity is warranted.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Copenhagen University, Copenhagen, Denmark.
| | - Tanni Borgbo
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Copenhagen University, Copenhagen, Denmark
| | - Søren Kløverpris
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
20
|
Signaroldi E, Laise P, Cristofanon S, Brancaccio A, Reisoli E, Atashpaz S, Terreni MR, Doglioni C, Pruneri G, Malatesta P, Testa G. Polycomb dysregulation in gliomagenesis targets a Zfp423-dependent differentiation network. Nat Commun 2016; 7:10753. [PMID: 26923714 PMCID: PMC4773478 DOI: 10.1038/ncomms10753] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Malignant gliomas constitute one of the most significant areas of unmet medical need, owing to the invariable failure of surgical eradication and their marked molecular heterogeneity. Accumulating evidence has revealed a critical contribution by the Polycomb axis of epigenetic repression. However, a coherent understanding of the regulatory networks affected by Polycomb during gliomagenesis is still lacking. Here we integrate transcriptomic and epigenomic analyses to define Polycomb-dependent networks that promote gliomagenesis, validating them both in two independent mouse models and in a large cohort of human samples. We find that Polycomb dysregulation in gliomagenesis affects transcriptional networks associated with invasiveness and de-differentiation. The dissection of these networks uncovers Zfp423 as a critical Polycomb-dependent transcription factor whose silencing negatively impacts survival. The anti-gliomagenic activity of Zfp423 requires interaction with the SMAD proteins within the BMP signalling pathway, pointing to a novel synergic circuit through which Polycomb inhibits BMP signalling.
Collapse
Affiliation(s)
- Elena Signaroldi
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Pasquale Laise
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Silvia Cristofanon
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Arianna Brancaccio
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Elisa Reisoli
- Trasferimento Genico, IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, Genoa 16132, Italy
| | - Sina Atashpaz
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Maria Rosa Terreni
- Pathology Department, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milan 20132, Italy
| | - Claudio Doglioni
- Pathology Department, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milan 20132, Italy
| | - Giancarlo Pruneri
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy
| | - Paolo Malatesta
- Trasferimento Genico, IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, Genoa 16132, Italy
- Department of Experimental Medicine (DiMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Via Festa del Perdono 7, Milan 20122, Italy
| |
Collapse
|
21
|
Kløverpris S, Mikkelsen JH, Pedersen JH, Jepsen MR, Laursen LS, Petersen SV, Oxvig C. Stanniocalcin-1 Potently Inhibits the Proteolytic Activity of the Metalloproteinase Pregnancy-associated Plasma Protein-A. J Biol Chem 2015. [PMID: 26195635 DOI: 10.1074/jbc.m115.650143] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stanniocalcin-1 (STC1) is a disulfide-bound homodimeric glycoprotein, first identified as a hypocalcemic hormone important for maintaining calcium homeostasis in teleost fish. STC1 was later found to be widely expressed in mammals, although it is not believed to function in systemic calcium regulation in these species. Several physiological functions of STC1 have been reported, although many molecular details are still lacking. We here demonstrate that STC1 is an inhibitor of the metzincin metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), which modulates insulin-like growth factor (IGF) signaling through proteolytic cleavage of IGF-binding proteins (IGFBPs). STC1 potently (Ki = 68 pm) inhibits PAPP-A cleavage of IGFBP-4, and we show in a cell-based assay that STC1 effectively antagonizes PAPP-A-mediated type 1 IGF receptor (IGF1R) phosphorylation. It has recently been found that the homologous STC2 inhibits PAPP-A proteolytic activity, and that this depends on the formation of a covalent complex between the inhibitor and the proteinase, mediated by Cys-120 of STC2. We find that STC1 is unable to bind covalently to PAPP-A, in agreement with the absence of a corresponding cysteine residue. It rather binds to PAPP-A with high affinity (KD = 75 pm). We further demonstrate that both STC1 and STC2 show inhibitory activity toward PAPP-A2, but not selected serine proteinases and metalloproteinases. We therefore conclude that the STCs are proteinase inhibitors, probably restricted in specificity to the pappalysin family of metzincin metalloproteinases. Our data are the first to identify STC1 as a proteinase inhibitor, suggesting a previously unrecognized function of STC1 in the IGF system.
Collapse
Affiliation(s)
| | | | | | | | | | - Steen V Petersen
- the Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Claus Oxvig
- From the Department of Molecular Biology and Genetics and
| |
Collapse
|
22
|
Engelmann JC, Amann T, Ott-Rötzer B, Nützel M, Reinders Y, Reinders J, Thasler WE, Kristl T, Teufel A, Huber CG, Oefner PJ, Spang R, Hellerbrand C. Causal Modeling of Cancer-Stromal Communication Identifies PAPPA as a Novel Stroma-Secreted Factor Activating NFκB Signaling in Hepatocellular Carcinoma. PLoS Comput Biol 2015; 11:e1004293. [PMID: 26020769 PMCID: PMC4447342 DOI: 10.1371/journal.pcbi.1004293] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/17/2015] [Indexed: 01/26/2023] Open
Abstract
Inter-cellular communication with stromal cells is vital for cancer cells. Molecules involved in the communication are potential drug targets. To identify them systematically, we applied a systems level analysis that combined reverse network engineering with causal effect estimation. Using only observational transcriptome profiles we searched for paracrine factors sending messages from activated hepatic stellate cells (HSC) to hepatocellular carcinoma (HCC) cells. We condensed these messages to predict ten proteins that, acting in concert, cause the majority of the gene expression changes observed in HCC cells. Among the 10 paracrine factors were both known and unknown cancer promoting stromal factors, the former including Placental Growth Factor (PGF) and Periostin (POSTN), while Pregnancy-Associated Plasma Protein A (PAPPA) was among the latter. Further support for the predicted effect of PAPPA on HCC cells came from both in vitro studies that showed PAPPA to contribute to the activation of NFκB signaling, and clinical data, which linked higher expression levels of PAPPA to advanced stage HCC. In summary, this study demonstrates the potential of causal modeling in combination with a condensation step borrowed from gene set analysis [Model-based Gene Set Analysis (MGSA)] in the identification of stromal signaling molecules influencing the cancer phenotype. All living cells rely on communication with other cells to ensure their function and survival. Molecular signals are sent among cells of the same cell type and from cells of one cell type to another. In cancer, not only the cancer cells themselves are responsible for the malignancy, but also stromal (non-cancerous) cells and the molecular signals they send to cancer cells are important factors that determine the severity and outcome of the disease. Therefore, the identification of stromal signals and their influence on cancer cells is important for the development of novel treatment strategies. With a computational systems biology model of stroma-cancer cell communication, we have compiled a set of ten proteins secreted by stromal cells that shape the cancer phenotype. Most importantly, our causal analysis uncovered Pregnancy-Associated Plasma Protein A (PAPPA) as a novel paracrine inducer of the pro-tumorigenic NFκB signaling pathway. In liver cancer patients, higher levels of PAPPA protein indicate a more progressed tumor stage, confirming its clinical relevance.
Collapse
Affiliation(s)
- Julia C. Engelmann
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| | - Thomas Amann
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Birgitta Ott-Rötzer
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Margit Nützel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Yvonne Reinders
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Jörg Reinders
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Wolfgang E. Thasler
- Biobank under the authority of Human Tissue and Cell Research (HTCR) and Center for Liver Cell Research, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Theresa Kristl
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg, Austria
| | - Andreas Teufel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Christian G. Huber
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg, Austria
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| |
Collapse
|
23
|
The role of PAPP-A in the IGF system: location, location, location. J Cell Commun Signal 2015; 9:177-87. [PMID: 25617049 DOI: 10.1007/s12079-015-0259-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
Although discovered as a placental protein present abundantly in the circulation of pregnant women, pregnancy-associated plasma protein-A (PAPP-A) is widely expressed in multiple tissues. PAPP-A is a highly specific metalloproteinase binding tightly to glycosaminoglycans present on the surface of cells. By cleaving a subset of insulin-like growth factor binding proteins (IGFBPs), PAPP-A thus functions within tissues as a growth-promoting enzyme, releasing bioactive IGF in close proximity to the IGF receptor. IGFBP-4 is believed to be the principal PAPP-A substrate, and the focus in this review is on PAPP-A enzymatic activity and its role in the PAPP-A-IGFBP-4-IGF axis, which is subject to regulation at several different levels. These include e.g., transcriptional control, competing reactions potentially sequestering IGF from IGFBP-4 and hence antagonizing PAPP-A-mediated IGF activation, and proteolytic inhibition of PAPP-A. The latter may involve the protein stanniocalcin-2 (STC2), recently found to potently inhibit PAPP-A activity by forming a covalent complex with PAPP-A. PAPP-A or complex-bound variants may escape from pathological tissues into the circulation. It is emphasized that the potential use of PAPP-A as a diagnostic or predictive biomarker in nonpregnant individuals requires precise knowledge of analyte identity and assay specificity in addition to an appropriate material for standardization. Finally, PAPP-A may serve as a therapeutic target to indirectly inhibit IGF signaling in tissues where this is driven by increased PAPP-A activity. By taking advantage of the intricate interaction between PAPP-A and IGFBP-4, highly specific and selective inhibition of PAPP-A is possible.
Collapse
|
24
|
Mansfield AS, Visscher DW, Hart SN, Wang C, Goetz MP, Oxvig C, Conover CA. Pregnancy-associated plasma protein-A expression in human breast cancer. Growth Horm IGF Res 2014; 24:264-267. [PMID: 25468445 PMCID: PMC4308469 DOI: 10.1016/j.ghir.2014.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/22/2014] [Accepted: 10/26/2014] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Pregnancy-associated plasma protein-A (PAPP-A) is a zinc metalloproteinase in the insulin-like growth factor system that is expressed by tissues outside of pregnancy and involved in normal and dysregulated growth. PAPP-A has been implicated in several cancers. However, studies of PAPP-A expression in breast cancer are limited. In this study, we assessed PAPP-A expression in different subtypes of human malignant breast cancer. Design Formalin-fixed paraffin-embedded tumor samples from 46 female patients with invasive breast cancer were divided into five defined groups [using markers for HER2, estrogen receptor, progesterone receptor, proliferation] that roughly correlate with molecularly defined subtypes (luminal A, luminal B, luminal/HER2 +, HER2 +, triple negative). These samples were analyzed for PAPP-A expression by immunohistochemistry. RESULTS PAPP-A staining in tumor tissue was detected in 45 of 46 specimens. There were significantly greater extent and intensity of PAPP-A expression in luminal B specimens with high proliferation index than luminal A specimens (P = 0.01). However, there were no differences between specimens positive or negative for HER2 (P = 0.14) or positive and negative for estrogen receptor (P = 0.31). CONCLUSION PAPP-A was detected in almost all breast cancer specimens and a more intense and greater extent of its expression was associated with luminal B specimens compared to luminal A specimens. The role of PAPP-A in breast cancer prognosis, and possibly therapeutics, warrants further investigation.
Collapse
Affiliation(s)
- Aaron S. Mansfield
- Division of Medical Oncology, Department of Laboratory Medicine, 200 First Street SW, Rochester, MN 55905 USA
| | | | - Steven N. Hart
- Health Sciences Research, 200 First Street SW, Rochester, MN 55905 USA
| | - Chen Wang
- Health Sciences Research, 200 First Street SW, Rochester, MN 55905 USA
| | - Matthew P. Goetz
- Division of Medical Oncology, Department of Laboratory Medicine, 200 First Street SW, Rochester, MN 55905 USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, MBG, Science Park, Gustav Wieds Vej 10C, DK-8000 Aarhus, Denmark
| | - Cheryl A. Conover
- Division of Endocrinology Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|