The Prognostic Value of ADAMTS8 and Its Role as a Tumor Suppressor in Breast Cancer

Abstract A disintegrin-like and metalloprotease with therombospondin type1 motif 8 (ADAMTS8) plays an important role in many malignancies. However, the clinical and biological significance of ADAMTS8 in breast cancer remain unknown. In this study, the clinical data from 1066 breast cancer patients were analyzed by The Cancer Genome Atlas (TCGA) database, and were analyzed using the correlation between ADAMTS8 expression and the clinicopathological features and prognoses. The CCK-8 assay, clone formation assay, flow cytometry and Transwell assay were used to characterize the effects of ADAMTS8 on proliferation, migration and invasion of breast cancer cells. Gene set enrichment analysis (GSEA) and western blotting were used to identify the potential molecular mechanism on how ADAMTS8 exert its biological function. ADAMTS8 overexpression correlated longer overall survival (OS) and progression-free survival (PFS). ADAMTS8 was considered as an independent prognostic factor for OS. ADAMTS8 overexpression inhibited breast cancer cell proliferation, migration and invasion in vitro, and induced G2/M cell cycle arrest. ADAMTS8 was also involved in cell cycle regulation and was associated with the EGFR/Akt signaling pathway. ADAMTS8 knockdown showed the reverse effect. Together, the results showed that ADAMTS8 functioned as a tumor suppressor gene (TGS) and could be a prognostic biomarker for breast cancer.

[1]  X. Lao,et al.  Global incidence and mortality of breast cancer: a trend analysis , 2021, Aging.

[2]  C. Frangou,et al.  USP1 Regulates TAZ Protein Stability Through Ubiquitin Modifications in Breast Cancer , 2020, Cancers.

[3]  Jianhua Wu,et al.  ADAMTS8 Inhibits Progression of Esophageal Squamous Cell Carcinoma. , 2020, DNA and cell biology.

[4]  M. Monteiro,et al.  Incomplete Pattern of Steroidogenic Protein Expression in Functioning Adrenocortical Carcinomas , 2020, Biomedicines.

[5]  T. Luo,et al.  ADAMTS8 is frequently down-regulated in colorectal cancer and functions as a tumor suppressor. , 2020, Biochemical and biophysical research communications.

[6]  Sung-Bae Kim,et al.  Prognostic value of tumor-infiltrating lymphocytes in patients with early-stage triple-negative breast cancers (TNBC) who did not receive adjuvant chemotherapy. , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[7]  Hao Wu,et al.  LINC00673 is activated by YY1 and promotes the proliferation of breast cancer cells via the miR-515-5p/MARK4/Hippo signaling pathway , 2019, Journal of Experimental & Clinical Cancer Research.

[8]  R. Xu,et al.  Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics? , 2019, Cancer communications.

[9]  Y. Jiao,et al.  High EIF2B5 mRNA expression and its prognostic significance in liver cancer: a study based on the TCGA and GEO database , 2018, Cancer management and research.

[10]  Jianhua Wu,et al.  ADAMTS8 targets ERK to suppress cell proliferation, invasion, and metastasis of hepatocellular carcinoma , 2018, OncoTargets and therapy.

[11]  G. Ren,et al.  PSMD2 regulates breast cancer cell proliferation and cell cycle progression by modulating p21 and p27 proteasomal degradation. , 2018, Cancer letters.

[12]  Q. Tao,et al.  The 3p14.2 tumour suppressor ADAMTS9 is inactivated by promoter CpG methylation and inhibits tumour cell growth in breast cancer , 2017, Journal of cellular and molecular medicine.

[13]  Lili Li,et al.  Epigenetic silencing of ADAMTS18 promotes cell migration and invasion of breast cancer through AKT and NF‐κB signaling , 2017, Cancer medicine.

[14]  A. Toker,et al.  AKT/PKB Signaling: Navigating the Network , 2017, Cell.

[15]  H. Takeuchi,et al.  Genetic and biochemical evidence that gastrulation defects in Pofut2 mutants result from defects in ADAMTS9 secretion. , 2016, Developmental biology.

[16]  Q. Tao,et al.  Protocadherin 17 functions as a tumor suppressor suppressing Wnt/β-catenin signaling and cell metastasis and is frequently methylated in breast cancer , 2016, Oncotarget.

[17]  M. Hung,et al.  Extracellular PKM2 induces cancer proliferation by activating the EGFR signaling pathway. , 2016, American journal of cancer research.

[18]  A. Appert-Collin,et al.  Role of ErbB Receptors in Cancer Cell Migration and Invasion , 2015, Front. Pharmacol..

[19]  Rui Henrique,et al.  Diagnostic and prognostic epigenetic biomarkers in cancer. , 2015, Epigenomics.

[20]  L. Aaltonen,et al.  Identification of homozygous deletion in ACAN and other candidate variants in familial classical Hodgkin lymphoma by exome sequencing , 2015, British journal of haematology.

[21]  Lian‐Wen Qi,et al.  Anti-Colon Cancer Effects of 6-Shogaol Through G2/M Cell Cycle Arrest by p53/p21-cdc2/cdc25A Crosstalk. , 2015, The American journal of Chinese medicine.

[22]  Jun Yu,et al.  MDGA2 is a novel tumour suppressor cooperating with DMAP1 in gastric cancer and is associated with disease outcome , 2015, Gut.

[23]  Zuli Yang,et al.  The roles of ADAMTS in angiogenesis and cancer , 2015, Tumor Biology.

[24]  Lili Li,et al.  Methylation of PLCD1 and adenovirus-mediated PLCD1 overexpression elicits a gene therapy effect on human breast cancer. , 2015, Experimental cell research.

[25]  Lirong Pei,et al.  Abstract 4012: Large-scale characterization of DNA methylation changes in human gastric carcinomas with and without metastasis , 2014 .

[26]  R. Roskoski The ErbB/HER family of protein-tyrosine kinases and cancer. , 2014, Pharmacological research.

[27]  Minghui Zhang,et al.  Prognostic value of survivin and EGFR protein expression in triple-negative breast cancer (TNBC) patients , 2014, Targeted Oncology.

[28]  Jun Yu,et al.  The Metalloprotease ADAMTS8 Displays Antitumor Properties through Antagonizing EGFR–MEK–ERK Signaling and Is Silenced in Carcinomas by CpG Methylation , 2013, Molecular Cancer Research.

[29]  A. Yee,et al.  The role of versican G3 domain in regulating breast cancer cell motility including effects on osteoblast cell growth and differentiation in vitro – evaluation towards understanding breast cancer cell bone metastasis , 2012, BMC Cancer.

[30]  John T. Wei,et al.  Beyond PSA: The Next Generation of Prostate Cancer Biomarkers , 2012, Science Translational Medicine.

[31]  A. Yee,et al.  Correction: Versican G3 Domain Modulates Breast Cancer Cell Apoptosis: A Mechanism for Breast Cancer Cell Response to Chemotherapy and EGFR Therapy , 2012, PLoS ONE.

[32]  Yosef Yarden,et al.  Feedback regulation of EGFR signalling: decision making by early and delayed loops , 2011, Nature Reviews Molecular Cell Biology.

[33]  Qiongqing Wang,et al.  ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. , 2009, Genes & development.

[34]  Samuel Leung,et al.  Basal-Like Breast Cancer Defined by Five Biomarkers Has Superior Prognostic Value than Triple-Negative Phenotype , 2008, Clinical Cancer Research.

[35]  Y. Okada,et al.  ADAMs in cancer cell proliferation and progression , 2007, Cancer science.

[36]  Farin Kamangar,et al.  Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[37]  R. Kerbel,et al.  Versican/PG‐M G3 domain promotes tumor growth and angiogenesis , 2004, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[38]  Sang-Oh Yoon,et al.  Roles of matrix metalloproteinases in tumor metastasis and angiogenesis. , 2003, Journal of biochemistry and molecular biology.

[39]  B. Hoffman,et al.  GADD45b and GADD45g are cdc2/cyclinB1 kinase inhibitors with a role in S and G2/M cell cycle checkpoints induced by genotoxic stress , 2002, Journal of cellular physiology.

[40]  M. Iruela-Arispe,et al.  METH-1, a Human Ortholog of ADAMTS-1, and METH-2 Are Members of a New Family of Proteins with Angio-inhibitory Activity* , 1999, The Journal of Biological Chemistry.