1
|
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: 0] [Impact Index Per Article: 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
|
2
|
Conover CA, Oxvig C. The Pregnancy-Associated Plasma Protein-A (PAPP-A) Story. Endocr Rev 2023; 44:1012-1028. [PMID: 37267421 DOI: 10.1210/endrev/bnad017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/01/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) was first identified in the early 1970s as a placental protein of unknown function, present at high concentrations in the circulation of pregnant women. In the mid-to-late 1990s, PAPP-A was discovered to be a metzincin metalloproteinase, expressed by many nonplacental cells, that regulates local insulin-like growth factor (IGF) activity through cleavage of high-affinity IGF binding proteins (IGFBPs), in particular IGFBP-4. With PAPP-A as a cell surface-associated enzyme, the reduced affinity of the cleavage fragments results in increased IGF available to bind and activate IGF receptors in the pericellular environment. This proteolytic regulation of IGF activity is important, since the IGFs promote proliferation, differentiation, migration, and survival in various normal and cancer cells. Thus, there has been a steady growth in investigation of PAPP-A structure and function outside of pregnancy. This review provides historical perspective on the discovery of PAPP-A and its structure and cellular function, highlights key studies of the first 50 years in PAPP-A research, and introduces new findings from recent years.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
3
|
Ruan P, Wang S, Yang C, Huang X, Sun P, Tan A. m 6A mRNA methylation regulates the ERK/NF-κB/AKT signaling pathway through the PAPPA/IGFBP4 axis to promote proliferation and tumor formation in endometrial cancer. Cell Biol Toxicol 2023; 39:1611-1626. [PMID: 35971034 DOI: 10.1007/s10565-022-09751-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
N6-methyladenosine (m6A) mRNA methylation has been considered a gene modulatory mechanism involved in disease progression and carcinogenesis. Herein, we aimed to explore the specific mechanism of m6A mRNA methylation in endometrial cancer. RT-qPCR was implemented to test the clinical correlation between m6A methylation and endometrial cancer. Bioinformatics analysis was performed to screen the genes related to endometrial cancer, and SRAMP was utilized for the prediction of m6A targets. Western blot assay and MeRIP-qPCR experiments were conducted to verify the effect of m6A methylation on the candidate genes and the signaling pathways involved in the occurrence of endometrial cancer. m6A-seq, RT-qPCR, and polysome profiling were used to confirm the mechanisms of m6A methylation in modulating related genes and pathways. The levels of m6A methylation, METTL3, and IGFBP4 were reduced in tumor tissues of patients with endometrial cancer, and SRAMP analysis confirmed that IGFBP4 and PAPPA had m6A methylation sites. Reduced m6A methylation promoted endometrial cancer cell progression and tumor formation in vivo. m6A methylation of RNA in endometrial cancer cells directly modulated IGFBP4 and PAPPA expression. m6A methylation regulated the PAPPA/IGFBP4 axis, thereby influencing endometrial cancer through the NF-κB and ERK signaling pathways. Knockdown of PAPPA or overexpression of IGFBP4 in endometrial cancer cells partially reduced disease progression caused by reduced m6A methylation. This research suggests that m6A mRNA methylation modulates the ERK/NF-κB/AKT signaling pathway through the PAPPA/IGFBP4 axis to induce cell proliferation and tumor formation in endometrial cancer. 1. METTL3 expressed modestly and m6A methylation of IGFBP4 and PAPPA mRNAs decreased in endometrial cancer; 2. YTHDF1-mediated IGFBP4 translation was reduced in HEC-1-A and AN3CA cells, and YTHDF2-mediated PAPPA mRNA degradation was blunted but its protein expression increased; 3. Increased PAPPA and reduced IGFBP4 activated IGF1-induced ERK, AKT, and NF-κB pathways by binding IGFR, thereby promoting cancer cell malignancy.
Collapse
Affiliation(s)
- Peng Ruan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Shujun Wang
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, 430060, People's Republic of China
| | - Chaoyi Yang
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, 430060, People's Republic of China
| | - Xiaohui Huang
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, 430060, People's Republic of China
| | - Pengxing Sun
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, 430060, People's Republic of China
| | - Aili Tan
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
4
|
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: 1.0] [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
|
5
|
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: 6] [Impact Index Per Article: 3.0] [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
|
6
|
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: 8] [Impact Index Per Article: 4.0] [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
|
7
|
Duan W, Wang K, Duan Y, Chen X, Chu X, Hu P, Xiong B. Combined Analysis of RNA Sequence and Microarray Data Reveals a Competing Endogenous RNA Network as Novel Prognostic Markers in Malignant Pleural Mesothelioma. Front Oncol 2021; 11:615234. [PMID: 33968720 PMCID: PMC8104912 DOI: 10.3389/fonc.2021.615234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a highly aggressive cancer with short survival time. Unbalanced competing endogenous RNAs (ceRNAs) have been shown to participate in the tumor pathogenesis and served as biomarkers for the clinical prognosis. However, the comprehensive analyses of the ceRNA network in the prognosis of MPM are still rarely reported. In this study, we obtained the transcriptome data of the MPM and the normal samples from TCGA, EGA, and GEO databases and identified the differentially expressed (DE) mRNAs, lncRNAs, and miRNAs. The functions of the prognostic genes and the overlapped DEmRNAs were further annotated by the multiple enrichment analyses. Then, the targeting relationships among lncRNA–miRNA and miRNA–mRNA were predicted and calculated, and a prognostic ceRNA regulatory network was established. We included the prognostic 73 mRNAs and 13 miRNAs and 26 lncRNAs into the ceRNA network. Moreover, 33 mRNAs, three miRNAs, and seven lncRNAs were finally associated with prognosis, and a model including seven mRNAs, two lincRNAs, and some clinical factors was finally established and validated by two independent cohorts, where CDK6 and SGMS1-AS1 were significant to be independent prognostic factors. In addition, the identified co-expressed modules associated with the prognosis were overrepresented in the ceRNA network. Multiple enrichment analyses showed the important roles of the extracellular matrix components and cell division dysfunction in the invasion of MPM potentially. In summary, the prognostic ceRNA network of MPM was established and analyzed for the first time and these findings shed light on the function of ceRNAs and revealed the potential prognostic and therapeutic biomarkers of MPM.
Collapse
Affiliation(s)
- Weicheng Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijie Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyi Chen
- Key Laboratory of Environment and Health (HUST), Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xufeng Chu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Hu
- Key Laboratory of Environment and Health (HUST), Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Xiong
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Cakiroglu E, Senturk S. Genomics and Functional Genomics of Malignant Pleural Mesothelioma. Int J Mol Sci 2020; 21:ijms21176342. [PMID: 32882916 PMCID: PMC7504302 DOI: 10.3390/ijms21176342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, aggressive cancer of the mesothelial cells lining the pleural surface of the chest wall and lung. The etiology of MPM is strongly associated with prior exposure to asbestos fibers, and the median survival rate of the diagnosed patients is approximately one year. Despite the latest advancements in surgical techniques and systemic therapies, currently available treatment modalities of MPM fail to provide long-term survival. The increasing incidence of MPM highlights the need for finding effective treatments. Targeted therapies offer personalized treatments in many cancers. However, targeted therapy in MPM is not recommended by clinical guidelines mainly because of poor target definition. A better understanding of the molecular and cellular mechanisms and the predictors of poor clinical outcomes of MPM is required to identify novel targets and develop precise and effective treatments. Recent advances in the genomics and functional genomics fields have provided groundbreaking insights into the genomic and molecular profiles of MPM and enabled the functional characterization of the genetic alterations. This review provides a comprehensive overview of the relevant literature and highlights the potential of state-of-the-art genomics and functional genomics research to facilitate the development of novel diagnostics and therapeutic modalities in MPM.
Collapse
Affiliation(s)
- Ece Cakiroglu
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
- Correspondence:
| |
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: 6] [Impact Index Per Article: 1.5] [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
|
Yu XH, He LH, Gao JH, Zhang DW, Zheng XL, Tang CK. Pregnancy-associated plasma protein-A in atherosclerosis: Molecular marker, mechanistic insight, and therapeutic target. Atherosclerosis 2018; 278:250-258. [DOI: 10.1016/j.atherosclerosis.2018.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/10/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
|
11
|
Smith YE, Toomey S, Napoletano S, Kirwan G, Schadow C, Chubb AJ, Mikkelsen JH, Oxvig C, Harmey JH. Recombinant PAPP-A resistant insulin-like growth factor binding protein 4 (dBP4) inhibits angiogenesis and metastasis in a murine model of breast cancer. BMC Cancer 2018; 18:1016. [PMID: 30348128 PMCID: PMC6196427 DOI: 10.1186/s12885-018-4950-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The Insulin-like growth factor (IGF) pathway plays a role in tumour development and progression. In vivo, IGF1 activity is regulated by the IGF binding proteins (IGFBPs). IGFBP4 inhibits the activity of IGF1 but proteolytic cleavage by pregnancy-associated plasma protein-A (PAPP-A) releases active IGF1. A modified IGFBP4, dBP4, which was resistant to PAPP-A cleavage but retained IGF1 binding capacity, was engineered, expressed in Human Embryonic Kidney (HEK) 293 cells and purified. This study examined the effects of dBP4 on IGF1-induced cell migration, invasion and angiogenesis in vitro. The effect of intra-tumour injections of dBP4 on tumour angiogenesis and metastasis was examined using the 4T1.2luc orthotopic model of breast cancer. METHODS PAPP-A resistance and IGF binding capacity of dBP4 were characterized by Western blot and surface plasmon resonance, respectively. 4T1.2luc are mouse mammary adenocarcinoma cells transfected with luciferase to allow in vivo imaging. The effect of dBP4 on IGF1-induced Akt activation in 4T1.2luc cells was assessed by Western blot. Cell migration and invasion assays were performed using 4T1.2luc cells. Angiokit™ assays and Matrigel® implants were used to assess the effects of dBP4 on angiogenesis in vitro and in vivo, respectively. An orthotopic breast cancer model - 4T1.2luc cells implanted in the mammary fat pad of BALB/c mice - was used to assess the effect of intra tumour injection of purified dBP4 on tumour angiogenesis and metastasis. Tumour growth and lung metastasis were examined by in vivo imaging and tumour angiogenesis was evaluated by CD31 immunohistochemistry. RESULTS Our engineered, PAPP-A resistant IGFBP4 (dBP4) retained IGF1 binding capacity and inhibited IGF1 activation of Akt as well as IGF1-induced migration and invasion by 4T1.2 mammary adenocarcinoma cells. dBP4 inhibited IGF1-induced angiogenesis in vitro and in Matrigel implants in vivo. Direct intra-tumour injection of soluble dBP4 reduced angiogenesis in 4T1.2 luc mammary tumours tumour and reduced lung metastasis. CONCLUSION A PAPP-A resistant IGFBP4, dBP4, inhibits angiogenesis and metastasis in 4T1.2 mammary fat pad tumours. This study highlights the therapeutic potential of dBP4 as an approach to block the tumour-promoting actions of IGF1.
Collapse
Affiliation(s)
- Y E Smith
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - S Toomey
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - S Napoletano
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - G Kirwan
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - C Schadow
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - A J Chubb
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - J H Mikkelsen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark
| | - C Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark
| | - J H Harmey
- Angiogenesis and Metastasis Research, Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
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: 7.3] [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
|
Guo Y, Bao Y, Guo D, Yang W. Pregnancy-associated plasma protein a in cancer: expression, oncogenic functions and regulation. Am J Cancer Res 2018; 8:955-963. [PMID: 30034934 PMCID: PMC6048405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023] Open
Abstract
Pregnancy-associated plasma protein A (PAPPA) is a protease that plays important roles in pregnancy, but interestingly acts as an oncogene outside of pregnancy. This review summarizes the oncogenic roles of PAPPA, including its expression levels in multiple malignancies, regulatory and signaling interactions, and pro-tumor functions, which include promoting tumor cell proliferation, invasion, migration and metastasis. These PAPPA activities are linked to IGFBP-4 proteolysis, increased IFG bioavailability, and activation of the NF-κB, PI3K/AKT and ERK signaling pathways. Therefore, PAPPA could be used as a biomarker for monitoring cancer development and progression as well as a potential therapeutic target.
Collapse
Affiliation(s)
- Yongchen Guo
- Institute of Precision Medicine, Jining Medical UniversityJining, China
| | - Yonghua Bao
- Institute of Precision Medicine, Jining Medical UniversityJining, China
| | - Dongli Guo
- Department of Surgery, Affiliated Hospital of Jining Medical UniversityJining, China
| | - Wancai Yang
- Institute of Precision Medicine, Jining Medical UniversityJining, China
- Department of Pathology, University of Illinois at ChicagoChicago, IL, USA
| |
Collapse
|
15
|
Bale LK, West SA, Conover CA. Inducible knockdown of pregnancy-associated plasma protein-A gene expression in adult female mice extends life span. Aging Cell 2017; 16:895-897. [PMID: 28600811 PMCID: PMC5506424 DOI: 10.1111/acel.12624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2017] [Indexed: 01/01/2023] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) knockout (KO) mice, generated through homologous recombination in embryonic stem cells, have a significantly increased lifespan compared to wild-type littermates. However, it is unknown whether this longevity advantage would pertain to PAPP-A gene deletion in adult animals. In the present study, we used tamoxifen (Tam)-inducible Cre recombinase-mediated excision of the floxed PAPP-A (fPAPP-A) gene in mice at 5 months of age. fPAPP-A mice, which were either positive (pos) or negative (neg) for Tam-Cre, received Tam treatment with quarterly boosters. Only female mice could be used with this experimental design. fPAPP-A/neg and fPAPP-A/pos mice had similar weights at the start of the experiment and showed equivalent weight gain. We found that fPAPP-A/pos mice had a significant extension of life span (P = 0.005). The median life span was increased by 21% for fPAPP-A/pos compared to fPAPP-A/neg mice. Analysis of mortality in life span quartiles indicated that the proportion of deaths of fPAPP-A/pos mice were lower than fPAPP-A/neg mice at young adult ages (P = 0.002 for 601-800 days) and higher than fPAPP-A/neg mice at older ages (P = 0.004 for >1000 days). Thus, survival curves and age-specific mortality indicate that female mice with knockdown of PAPP-A gene expression as adults have an extended healthy life span.
Collapse
Affiliation(s)
- Laurie K. Bale
- Endocrine Research UnitMayo Clinic200 First Street SWRochesterMN55905USA
| | - Sally A. West
- Endocrine Research UnitMayo Clinic200 First Street SWRochesterMN55905USA
| | - Cheryl A. Conover
- Endocrine Research UnitMayo Clinic200 First Street SWRochesterMN55905USA
| |
Collapse
|
16
|
Conover CA, Oxvig C. PAPP-A: a promising therapeutic target for healthy longevity. Aging Cell 2017; 16:205-209. [PMID: 28035757 PMCID: PMC5334524 DOI: 10.1111/acel.12564] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2016] [Indexed: 12/16/2022] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) is a proteolytic enzyme that was discovered to increase local insulin-like growth factor (IGF) availability for receptor activation through cleavage of inhibitory IGF binding proteins (IGFBPs). Reduced IGF signaling has been associated with increased lifespan and healthspan. Therefore, inhibition of PAPP-A represents a novel approach to indirectly decrease the availability of bioactive IGF. Here, we will review data in support of PAPP-A as a therapeutic target to promote healthy longevity.
Collapse
Affiliation(s)
| | - Claus Oxvig
- Department of Molecular Biology and Genetics; Aarhus Universitet; Aarhus Denmark
| |
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: 3.6] [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
|
Li X, Liu J, Zhang Y. Octylphenol induced gene expression in testes of Frog, Rana chensinensis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2016; 128:75-82. [PMID: 26896894 DOI: 10.1016/j.ecoenv.2016.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 02/11/2016] [Accepted: 02/11/2016] [Indexed: 06/05/2023]
Abstract
Octylphenol (OP) is an endocrine-disrupting chemical (EDC), which can disrupt the reproductive system. To understand the effect of OP, a subtractive cDNA library was constructed using suppression subtractive hybridization (SSH) to identify alterations of gene transcription in the testes of the frog Rana chensinensis after OP exposure. Two hundred positive clones were selected and 134 sequences of gene fragments were produced from the subtractive library randomly. These genes were identified to be involved in metabolic process, cellular process, biological regulation, stimulus, immune system and female pregnancy process. In order to verify the efficiency of the subtractive cDNA library, PSG9 and PAPP-A were analyzed further as two representatives of differentially expressed transcription genes using semi-quantitative RT-PCR. Our result was the first successful construction of the subtractive cDNA library in frog testes after OP treatment. Based on this cDNA library, OP was shown to affect multiple physiological processes including inducing immune response, disrupting the steroid hormone synthesis and influencing spermatogenesis in the testis by up-regulation of specific genes.
Collapse
Affiliation(s)
- Xinyi Li
- Co-Innovation Center for Qinba Regions'Sustainable Development, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jia Liu
- Co-Innovation Center for Qinba Regions'Sustainable Development, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yuhui Zhang
- Co-Innovation Center for Qinba Regions'Sustainable Development, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China.
| |
Collapse
|
19
|
Prithviraj P, Anaka M, McKeown SJ, Permezel M, Walkiewicz M, Cebon J, Behren A, Jayachandran A. Pregnancy associated plasma protein-A links pregnancy and melanoma progression by promoting cellular migration and invasion. Oncotarget 2016; 6:15953-65. [PMID: 25940796 PMCID: PMC4599249 DOI: 10.18632/oncotarget.3643] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 03/23/2015] [Indexed: 11/25/2022] Open
Abstract
Melanoma is the most common cancer diagnosed in pregnant women and an aggressive course with poorer outcomes is commonly described during pregnancy or shortly after childbirth. The underlying mechanisms for this are not understood. Here, we report that melanoma migration, invasiveness and progression are promoted by pregnancy-associated plasma protein-A (PAPPA), a pregnancy-associated metalloproteinase produced by the placenta that increases the bioavailability of IGF1 by cleaving it from a circulating complex formed with IGFBP4. We show that PAPPA is widely expressed by metastatic melanoma tumors and is elevated in melanoma cells exhibiting mesenchymal, invasive and label-retaining phenotypes. Notably, inhibition of PAPPA significantly reduced invasion and migration of melanoma cells in vitro and in vivo within the embryonic chicken neural tube. PAPPA-enriched pregnancy serum treatment enhanced melanoma motility in vitro. Furthermore, we report that IGF1 can induce the phenotypic and functional effects of epithelial-to-mesenchymal transition (EMT) in melanoma cells. In this study, we establish a clear relationship between a pregnancy-associated protein PAPPA, melanoma and functional effects mediated through IGF1 that provides a plausible mechanism for accelerated melanoma progression during pregnancy. This opens the possibility of targeting the PAPPA/IGF1 axis therapeutically.
Collapse
Affiliation(s)
- Prashanth Prithviraj
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia
| | - Matthew Anaka
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia
| | - Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, VIC, Australia
| | | | - Marzena Walkiewicz
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia
| | - Jonathan Cebon
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| | - Andreas Behren
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| | - Aparna Jayachandran
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Olivia Newton-John Cancer and Wellness Centre, Heidelberg, VIC, Australia.,Department of Medicine, University of Melbourne, VIC, Australia.,School of Cancer Medicine, La Trobe University, VIC, Australia
| |
Collapse
|
20
|
Mikkelsen JH, Resch ZT, Kalra B, Savjani G, Kumar A, Conover CA, Oxvig C. Indirect targeting of IGF receptor signaling in vivo by substrate-selective inhibition of PAPP-A proteolytic activity. Oncotarget 2015; 5:1014-25. [PMID: 24572990 PMCID: PMC4011579 DOI: 10.18632/oncotarget.1629] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The insulin-like growth factor (IGF) signaling pathway is involved in certain human cancers, and the feasibility of directly targeting the IGF receptor has been actively investigated. However, recent evidence from clinical trials suggests that this approach can be problematic. We have developed an alternative strategy to indirectly inhibit the IGF signaling by targeting the metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A). PAPP-A associated with the cell surface cleaves IGF binding protein-4 (IGFBP-4), when IGF is bound to IGFBP-4, and thereby increases IGF bioavailability for receptor activation in an autocrine/paracrine manner. We hypothesized that inhibition of PAPP-A would suppress excessive local IGF signaling in tissues where this is caused by increased PAPP-A proteolytic activity. To test this hypothesis, we developed an inhibitory monoclonal antibody, mAb 1/41, which targets a unique substrate-binding exosite of PAPP-A. This inhibitor selectively and specifically inhibits proteolytic cleavage of IGFBP-4 with an inhibitory constant (Ki) of 135 pM. In addition, it inhibited intracellular signaling of the IGF receptor (AKT phosphorylation) in monolayers of A549 cells, an IGF-responsive lung cancer-derived cell line found to express high levels of PAPP-A. We further showed that mAb 1/41 is effective towards PAPP-A bound to cell surfaces, and that it is capable of inhibiting PAPP-A activity in vivo. Using a murine xenograft model of A549 cells, we demonstrated that mAb 1/41 administered intraperitoneally significantly inhibited tumor growth. Analysis of xenograft tumor tissue recovered from treated mice showed penetration of mAb 1/41, reduced IGFBP-4 proteolysis, and reduced AKT phosphorylation. Our study provides proof of concept that IGF signaling can be selectively reduced by targeting a regulatory proteinase that functions extracellularly, upstream of the IGF receptor. PAPP-A targeting thus represents an alternative therapeutic strategy for inhibiting IGF receptor signaling.
Collapse
Affiliation(s)
- Jakob H Mikkelsen
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
21
|
Becker MA, Haluska P, Bale LK, Oxvig C, Conover CA. A novel neutralizing antibody targeting pregnancy-associated plasma protein-a inhibits ovarian cancer growth and ascites accumulation in patient mouse tumorgrafts. Mol Cancer Ther 2015; 14:973-81. [PMID: 25695953 DOI: 10.1158/1535-7163.mct-14-0880] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/10/2015] [Indexed: 12/16/2022]
Abstract
The majority of ovarian cancer patients acquire resistance to standard platinum chemotherapy and novel therapies to reduce tumor burden and ascites accumulation are needed. Pregnancy-associated plasma protein-A (PAPP-A) plays a key role in promoting insulin-like growth factor (IGF) pathway activity, which directly correlates to ovarian cancer cell transformation, growth, and invasiveness. Herein, we evaluate PAPP-A expression in tumors and ascites of women with ovarian cancer, and determine the antitumor efficacy of a neutralizing monoclonal PAPP-A antibody (mAb-PA) in ovarian cancer using primary patient ovarian tumorgrafts ("Ovatars"). PAPP-A mRNA expression in patient ovarian tumors correlated with poor outcome and was validated as a prognostic surrogate in Ovatar tumors. Following confirmation of mAb-PA bioavailability and target efficacy in vivo, the antitumor efficacy of mAb-PA in multiple Ovatar tumor models was examined and the response was found to depend on PAPP-A expression. Strikingly, the addition of mAb-PA to standard platinum chemotherapy effectively sensitized platinum-resistant Ovatar tumors. PAPP-A protein in ascites was also assessed in a large cohort of patients and very high levels were evident across the entire sample set. Therefore, we evaluated targeted PAPP-A inhibition as a novel approach to managing ovarian ascites, and found that mAb-PA inhibited the development, attenuated the progression, and induced the regression of Ovatar ascites. Together, these data indicate PAPP-A as a potential palliative and adjunct therapeutic target for women with ovarian cancer.
Collapse
Affiliation(s)
- Marc A Becker
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Paul Haluska
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Laurie K Bale
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
22
|
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: 133] [Impact Index Per Article: 14.8] [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
|
23
|
Crosley EJ, Dunk CE, Beristain AG, Christians JK. IGFBP-4 and -5 are expressed in first-trimester villi and differentially regulate the migration of HTR-8/SVneo cells. Reprod Biol Endocrinol 2014; 12:123. [PMID: 25475528 PMCID: PMC4271501 DOI: 10.1186/1477-7827-12-123] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/26/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adverse gestational outcomes such as preeclampsia (PE) and intrauterine growth restriction (IUGR) are associated with placental insufficiency. Normal placental development relies on the insulin-like growth factors -I and -II (IGF-I and -II), in part to stimulate trophoblast proliferation and extravillous trophoblast (EVT) migration. The insulin-like growth factor binding proteins (IGFBPs) modulate the bioavailability of IGFs in various ways, including sequestration, potentiation, and/or increase in half-life. The roles of IGFBP-4 and -5 in the placenta are unknown, despite consistent associations between pregnancy complications and the levels of two IGFBP-4 and/or -5 proteases, pregnancy-associated plasma protein -A and -A2 (PAPP-A and PAPP-A2). The primary objective of this study was to elucidate the effects of IGFBP-4 and -5 on IGF-I and IGF-II in a model of EVT migration. A related objective was to determine the timing and location of IGFBP-4 and -5 expression in the placental villi. METHODS We used wound healing assays to examine the effects of IGFBP-4 and -5 on the migration of HTR-8/SVneo cells following 4 hours of serum starvation and 24 hours of treatment. Localization of IGFBP-4, -5 and PAPP-A2 was assessed by immunohistochemical staining of first trimester placental sections. RESULTS 2 nM IGF-I and -II each increased HTR-8/SVneo cell migration with IGF-I increasing migration significantly more than IGF-II. IGFBP-4 and -5 showed different levels of inhibition against IGF-I. 20 nM IGFBP-4 completely blocked the effects of 2 nM IGF-I, while 20 nM IGFBP-5 significantly reduced the effects of 2 nM IGF-I, but not to control levels. Either 20 nM IGFBP-4 or 20 nM IGFBP-5 completely blocked the effects of 2 nM IGF-II. Immunohistochemistry revealed co-localization of IGFBP-4, IGFBP-5 and PAPP-A2 in the syncytiotrophoblast layer of first trimester placental villi as early as 5 weeks of gestational age. CONCLUSIONS IGFBP-4 and -5 show different levels of inhibition on the migration-stimulating effects of IGF-I and IGF-II, suggesting different roles for PAPP-A and PAPP-A2. Moreover, co-localization of the pappalysins and their substrates within placental villi suggests undescribed roles of these molecules in early placental development.
Collapse
Affiliation(s)
- Erin J Crosley
- Biological Sciences, Simon Fraser University, V5A 1S6 Burnaby, Canada
| | - Caroline E Dunk
- Research Centre for Women’s and Infants Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Alexander G Beristain
- Department of Obstetrics and Gynecology, The University of British Columbia, Vancouver, Canada
- The Child and Family Research Institute, Vancouver, Canada
| | | |
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: 28] [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
|
25
|
Mikkelsen JH, Steffensen LB, Oxvig C. Development of a recombinant antibody towards PAPP-A for immunohistochemical use in multiple animal species. J Immunol Methods 2013; 404:33-40. [PMID: 24333852 DOI: 10.1016/j.jim.2013.12.002] [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: 11/04/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
The metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), is increasingly recognized as a modulator of insulin-like growth factor (IGF) signaling; it cleaves IGF binding proteins causing the release of bioactive IGF. Accumulating evidence supports an important role of PAPP-A in both normal physiology and under different pathological conditions. However, antibodies for the detection of PAPP-A in non-human tissues have been lacking, although needed for use with several animal models which are currently being developed. To develop a monoclonal antibody suitable for the immunohistochemical detection of PAPP-A, we therefore selected a phage-derived scFv antibody, PAC1, specifically recognizing an epitope of PAPP-A, which is highly conserved between multiple animal species. We first converted this antibody into bivalent IgG, and verified its ability to recognize PAPP-A in sections of formalin-fixed and paraffin-embedded tissue. For increased sensitivity, affinity maturation to sub-nanomolar affinity was then carried out. The resulting recombinant antibody, PAC1-D8-mIgG2a, detects PAPP-A specifically and sensitively in human tissue. In addition, this antibody allows detection of PAPP-A in non-human species. We demonstrate its usefulness for the visualization of PAPP-A in murine and porcine tissues.
Collapse
Affiliation(s)
- Jakob H Mikkelsen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Lasse B Steffensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark.
| |
Collapse
|