1
|
Han L, Chen S, Du SY. Role of inositol polyphosphate-4-phosphatase type II in oncogenesis of digestive system tumors. World J Gastrointest Oncol 2023; 15:1706-1716. [PMID: 37969410 PMCID: PMC10631434 DOI: 10.4251/wjgo.v15.i10.1706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/22/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
Inositol polyphosphate-4-phosphatase type II (INPP4B) is a newly discovered PI(3,4,5)P3 phosphatase. Many studies have revealed that INPP4B is upregulated or downregulated in tumors of the digestive system, and the abnormal expression of INPP4B may be attributed to the occurrence, development, and prognosis of tumors of the digestive system. This paper reviews studies on the correlations between INPP4B and digestive system tumors and the roles of INPP4B in the development of different tumors to provide a theoretical basis for further research on its molecular mechanism and clinical application. "INPP4B" and "tumor" were searched as key words in PubMed and in the CNKI series full text database retrieval system from January 2000 to August 2023. A total of 153 English-language studies and 30 Chinese-language studies were retrieved. The following enrollment criteria were applied: (1) Studies contained information on the biological structure and functions of INPP4B; (2) studies covered the influence of abnormal expression of INPP4B in digestive system tumors; and (3) studies covered the role of INPP4B in the diagnosis, treatment, and prognosis of digestive system tumors. After excluding the literature irrelevant to this study, 61 papers were finally included in the analysis. INPP4B expression is low in gastric cancer, colon cancer, pancreatic cancer, and liver cancer but it has high expression in esophageal cancer, colon cancer, pancreatic cancer, and gallbladder cancer. INPP4B is involved in the occurrence and development of digestive system tumors through the regulation of gene expression and signal transduction. The abnormal expression of INPP4B plays an important role in the development of digestive system tumors. Studies on INPP4B provide new molecular insights for the diagnosis, treatment, and prognosis evaluation of digestive system tumors.
Collapse
Affiliation(s)
- Le Han
- Peking University China-Japan Friendship School of Clinical Medicine, Peking University, Beijing 100029, China
| | - Shuo Chen
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shi-Yu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
2
|
Huang J, Chen L, Wu J, Ai D, Zhang JQ, Chen TG, Wang L. Targeting the PI3K/AKT/mTOR Signaling Pathway in the Treatment of Human Diseases: Current Status, Trends, and Solutions. J Med Chem 2022; 65:16033-16061. [PMID: 36503229 DOI: 10.1021/acs.jmedchem.2c01070] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway is one of the most important intracellular pathways involved in cell proliferation, growth, differentiation, and survival. Therefore, this route is a prospective biological target for treating various human diseases, such as tumors, neurodegenerative diseases, pulmonary fibrosis, and diabetes. An increasing number of clinical studies emphasize the necessity of developing novel molecules targeting the PI3K/AKT/mTOR pathway. This review focuses on recent advances in ATP-competitive inhibitors, allosteric inhibitors, covalent inhibitors, and proteolysis-targeting chimeras against the PI3K/AKT/mTOR pathway, and highlights possible solutions for overcoming the toxicities and acquired drug resistance of currently available drugs. We also provide recommendations for the future design and development of promising drugs targeting this pathway.
Collapse
Affiliation(s)
- Jindi Huang
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Liye Chen
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiangxia Wu
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Daiqiao Ai
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ji-Quan Zhang
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Tie-Gen Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Room 109, Building C, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Ling Wang
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
3
|
Mazumdar A, Tahaney WM, Hill JL, Zhang Y, Ramachandran S, Kawedia J, Qian J, Contreras A, Savage MI, Vornik LA, Sei S, Mohammed A, Brown PH. Targeting the mTOR Pathway for the Prevention of ER-Negative Breast Cancer. Cancer Prev Res (Phila) 2022; 15:791-802. [PMID: 35981902 PMCID: PMC9762336 DOI: 10.1158/1940-6207.capr-22-0106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/18/2022] [Accepted: 08/15/2022] [Indexed: 01/31/2023]
Abstract
PREVENTION RELEVANCE Our results show that everolimus delays mammary tumor formation in multiple mouse models, suggesting that mTOR inhibitors will be useful for the prevention of ER-negative and triple-negative breast cancer in humans. See related Spotlight, p. 787.
Collapse
Affiliation(s)
- Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Corresponding Author: Abhijit Mazumdar, Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030. E-mail:
| | - William M. Tahaney
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jamal L. Hill
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yun Zhang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sumankalai Ramachandran
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jitesh Kawedia
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Qian
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Contreras
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michelle I. Savage
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lana A. Vornik
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shizuko Sei
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Altaf Mohammed
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
4
|
Bennett C, Carroll C, Wright C, Awad B, Park JM, Farmer M, Brown E(B, Heatherly A, Woodard S. Breast Cancer Genomics: Primary and Most Common Metastases. Cancers (Basel) 2022; 14:3046. [PMID: 35804819 PMCID: PMC9265113 DOI: 10.3390/cancers14133046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Specific genomic alterations have been found in primary breast cancer involving driver mutations that result in tumorigenesis. Metastatic breast cancer, which is uncommon at the time of disease onset, variably impacts patients throughout the course of their disease. Both the molecular profiles and diverse genomic pathways vary in the development and progression of metastatic breast cancer. From the most common metastatic site (bone), to the rare sites such as orbital, gynecologic, or pancreatic metastases, different levels of gene expression indicate the potential involvement of numerous genes in the development and spread of breast cancer. Knowledge of these alterations can, not only help predict future disease, but also lead to advancement in breast cancer treatments. This review discusses the somatic landscape of breast primary and metastatic tumors.
Collapse
Affiliation(s)
- Caroline Bennett
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Caleb Carroll
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Cooper Wright
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Barbara Awad
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN 37752, USA;
| | - Jeong Mi Park
- Department of Radiology, The University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA;
| | - Meagan Farmer
- Department of Genetics, Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL 35233, USA; (M.F.); (A.H.)
| | - Elizabeth (Bryce) Brown
- Laboratory Genetics Counselor, UAB Medical Genomics Laboratory, Kaul Human Genetics Building, 720 20th Street South, Suite 332, Birmingham, AL 35294, USA;
| | - Alexis Heatherly
- Department of Genetics, Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL 35233, USA; (M.F.); (A.H.)
| | - Stefanie Woodard
- Department of Radiology, The University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA;
| |
Collapse
|
5
|
Asleh K, Tu D, Gao D, Bramwell V, Levine MN, Pritchard KI, Shepherd LE, Nielsen TO. Predictive Significance of an Optimized Panel for Basal-like Breast Cancer: Results from the Canadian Cancer Trials Group MA.5 and MA.12 Phase III Clinical Trials. Clin Cancer Res 2021; 27:6570-6579. [PMID: 34615722 DOI: 10.1158/1078-0432.ccr-21-1942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Accurate IHC biomarkers incorporating nestin positivity or inositol polyphosphate-4-phosphate (INPP4B) loss have recently been optimized to identify the basal-like intrinsic breast cancer subtype regardless of estrogen, progesterone, or Her2 status. We examined the predictive capacity of these basal biomarkers in the CCTG MA.5 chemotherapy and MA.12 endocrine therapy trials. EXPERIMENTAL DESIGN Formalin-fixed paraffin embedded blocks of primary tumors from patients randomized in the two trials were used to build tissue microarrays. IHC staining for nestin and INPP4B followed published methods and REMARK criteria. A prespecified statistical plan tested the hypothesis that patients with basal breast cancer (nestin+ or INPP4B-) would not benefit from anthracycline substitution in MA.5 or from tamoxifen in MA.12. RESULTS Nestin positivity or INPP4B loss was observed in 110/453 (24%) interpretable samples from MA.5 and 47/366 (13%) from MA.12, and was associated with high grade, younger age, estrogen receptor negativity, triple-negative, core basal, and PAM50 basal-like subtypes. In the MA.5 trial, patients assigned as basal experienced lower benefit from anthracycline versus nonanthracycline adjuvant chemotherapy [HR, 1.49; 95% confidence interval (CI), 0.72-3.10] when compared with non-basal (nestin- and INPP4B+) cases where there was a higher benefit from anthracyclines (HR, 0.75; 95% CI, 0.54-1.04; P interaction = 0.01). In the MA.12 trial, patients assigned as basal did not demonstrate a benefit from adjuvant tamoxifen versus placebo (HR, 0.48; 95% CI, 0.12-1.86; P = 0.29), whereas nonbasal cases displayed significant benefit (HR, 0.66; 95% CI, 0.45-0.98; P = 0.04), although the interaction test was not significant. CONCLUSIONS The nestin/INPP4B IHC panel identifies women with basal breast cancers who benefit from nonanthracycline chemotherapy but not endocrine adjuvant treatments.
Collapse
Affiliation(s)
- Karama Asleh
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vivien Bramwell
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Mark N Levine
- Department of Medical Oncology, McMaster University, Hamilton, Ontario, Canada
| | | | - Lois E Shepherd
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Torsten O Nielsen
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
6
|
Targeting SHIP1 and SHIP2 in Cancer. Cancers (Basel) 2021; 13:cancers13040890. [PMID: 33672717 PMCID: PMC7924360 DOI: 10.3390/cancers13040890] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Phosphoinositol signaling pathways and their dysregulation have been shown to have a fundamental role in health and disease, respectively. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, are regulators of the PI3K/AKT pathway that have crucial roles in cancer progression. This review aims to summarize the role of SHIP1 and SHIP2 in cancer signaling and the immune response to cancer, the discovery and use of SHIP inhibitors and agonists as possible cancer therapeutics. Abstract Membrane-anchored and soluble inositol phospholipid species are critical mediators of intracellular cell signaling cascades. Alterations in their normal production or degradation are implicated in the pathology of a number of disorders including cancer and pro-inflammatory conditions. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, play a fundamental role in these processes by depleting PI(3,4,5)P3, but also by producing PI(3,4)P2 at the inner leaflet of the plasma membrane. With the intent of targeting SHIP1 or SHIP2 selectively, or both paralogs simultaneously, small molecule inhibitors and agonists have been developed and tested in vitro and in vivo over the last decade in various disease models. These studies have shown promising results in various pre-clinical models of disease including cancer and tumor immunotherapy. In this review the potential use of SHIP inhibitors in cancer is discussed with particular attention to the molecular structure, binding site and efficacy of these SHIP inhibitors.
Collapse
|
7
|
Tran KB, Kolekar S, Jabed A, Jaynes P, Shih JH, Wang Q, Flanagan JU, Rewcastle GW, Baguley BC, Shepherd PR. Diverse mechanisms activate the PI 3-kinase/mTOR pathway in melanomas: implications for the use of PI 3-kinase inhibitors to overcome resistance to inhibitors of BRAF and MEK. BMC Cancer 2021; 21:136. [PMID: 33549048 PMCID: PMC7866738 DOI: 10.1186/s12885-021-07826-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background The PI 3-kinase (PI3K) pathway has been implicated as a target for melanoma therapy. Methods Given the high degree of genetic heterogeneity in melanoma, we sought to understand the breadth of variation in PI3K signalling in the large NZM panel of early passage cell lines developed from metastatic melanomas. Results We find the vast majority of lines show upregulation of this pathway, and this upregulation is achieved by a wide range of mechanisms. Expression of all class-IA PI3K isoforms was readily detected in these cell lines. A range of genetic changes in different components of the PI3K pathway was seen in different lines. Coding variants or amplification were identified in the PIK3CA gene, and amplification of the PK3CG gene was common. Deletions in the PIK3R1 and PIK3R2 regulatory subunits were also relatively common. Notably, no genetic variants were seen in the PIK3CD gene despite p110δ being expressed in many of the lines. Genetic variants were detected in a number of genes that encode phosphatases regulating the PI3K signalling, with reductions in copy number common in PTEN, INPP4B, INPP5J, PHLLP1 and PHLLP2 genes. While the pan-PI3K inhibitor ZSTK474 attenuated cell growth in all the lines tested, isoform-selective inhibition of p110α and p110δ inhibited cell growth in only a subset of the lines and the inhibition was only partial. This suggests that functional redundancy exists between PI3K isoforms. Furthermore, while ZSTK474 was initially effective in melanoma cells with induced resistance to vemurafenib, a subset of these cell lines concurrently developed partial resistance to PI3K inhibition. Importantly, mTOR-selective or mTOR/PI3K dual inhibitors effectively inhibited cell growth in all the lines, including those already resistant to BRAF inhibitors and ZSTK474. Conclusions Overall, this indicates a high degree of diversity in the way the PI3K pathway is activated in different melanoma cell lines and that mTOR is the most effective point for targeting the growth via the PI3K pathway across all of these cell lines. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07826-4.
Collapse
Affiliation(s)
- Khanh B Tran
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Sharada Kolekar
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anower Jabed
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Patrick Jaynes
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Jen-Hsing Shih
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Qian Wang
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Jack U Flanagan
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Gordon W Rewcastle
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Bruce C Baguley
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand. .,Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
8
|
Zhao X, Jiang Y, Jiang T, Han X, Wang Y, Chen L, Feng X. Physiological and pathological regulation of autophagy in pregnancy. Arch Gynecol Obstet 2020; 302:293-303. [PMID: 32556514 DOI: 10.1007/s00404-020-05607-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/18/2020] [Indexed: 12/19/2022]
Abstract
Autophagy exists widely in eukaryotic cells and is regulated by a variety of molecular mechanisms. Its physiological functions include providing energy, maintaining cell homeostasis, and promoting apoptosis of abnormal cells. At present, the regulation of autophagy in tumor, degenerative disease, and cardiovascular disease has attracted much attention. Gradually, the role of autophagy in pregnancy tends to be valued. The previous literature has shown that autophagy can influence the occurrence and maintenance of pregnancy from three aspects: embryo (affecting the process of fertilization and embryonic development and the function of trophoblast cells), maternal (decidualization), and maternal-to-fetal immune crosstalk. Undoubtedly, abnormalities in autophagy levels are associated with a variety of pregnancy complications, such as preeclampsia, fetal growth restriction, and preterm delivery which have been proven by human, animal, and in vitro experiments. The regulation of autophagy is expected to be a target for the treatment of these pregnancy complications. This article reviews the research on autophagy, especially about its physiological and pathological regulation during pregnancy.
Collapse
Affiliation(s)
- Xiaoxuan Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yuepeng Jiang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Tianyue Jiang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xinyu Han
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Ying Wang
- Department of First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Lu Chen
- Department of First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xiaoling Feng
- Department of First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
9
|
Using Phosphatidylinositol Phosphorylation as Markers for Hyperglycemic Related Breast Cancer. Int J Mol Sci 2020; 21:ijms21072320. [PMID: 32230859 PMCID: PMC7177416 DOI: 10.3390/ijms21072320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Studies have suggested that type 2 diabetes (T2D) is associated with a higher incidence of breast cancer and related mortality rates. T2D postmenopausal women have an ~20% increased chance of developing breast cancer, and women with T2D and breast cancer have a 50% increase in mortality compared to breast cancer patients without diabetes. This correlation has been attributed to the general activation of insulin receptor signaling, glucose metabolism, phosphatidylinositol (PI) kinases, and growth pathways. Furthermore, the presence of breast cancer specific PI kinase and/or phosphatase mutations enhance metastatic breast cancer phenotypes. We hypothesized that each of the breast cancer subtypes may have characteristic PI phosphorylation profiles that are changed in T2D conditions. Therefore, we sought to characterize the PI phosphorylation when equilibrated in normal glycemic versus hyperglycemic serum conditions. Our results suggest that hyperglycemia leads to: 1) A reduction in PI3P and PIP3, with increased PI4P that is later converted to PI(3,4)P2 at the cell surface in hormone receptor positive breast cancer; 2) a reduction in PI3P and PI4P with increased PIP3 surface expression in human epidermal growth factor receptor 2-positive (HER2+) breast cancer; and 3) an increase in di- and tri-phosphorylated PIs due to turnover of PI3P in triple negative breast cancer. This study begins to describe some of the crucial changes in PIs that play a role in T2D related breast cancer incidence and metastasis.
Collapse
|
10
|
Yang Q, Li H, Xiao Y, Wu C, Yang S, Sun Z. Expression of inositol polyphosphate 4‐phosphatase type II and the prognosis of oral squamous cell carcinoma. Eur J Oral Sci 2020; 128:37-45. [PMID: 32027770 DOI: 10.1111/eos.12673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Qi‐Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Hao Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Yao Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Cong‐Cong Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Shao‐Chen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - Zhi‐Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral Maxillofacial‐Head Neck Oncology School & Hospital of Stomatology Wuhan University Wuhan China
| |
Collapse
|
11
|
Darbeheshti F, Rezaei N, Amoli MM, Mansoori Y, Tavakkoly Bazzaz J. Integrative analyses of triple negative dysregulated transcripts compared with non-triple negative tumors and their functional and molecular interactions. J Cell Physiol 2019; 234:22386-22399. [PMID: 31081218 DOI: 10.1002/jcp.28804] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
Abstract
Triple-negative (TN) tumors are a subtype of breast cancer with aggressive behaviors and limited targeted therapies. Microarray studies were not concerned with interactions and functional relations of dysregulated transcripts. Here, we aimed to conduct integrative strategy to analyze gene and miRNA available microarray data as well as bioinformatic analyses to catch a more inclusive picture of pivotal dysregulated transcripts and their interactions in TN tumors. Several online datasets and offline bioinformatic tools were used to detect differentially expressed (DE) transcripts, both protein and nonprotein coding, in TN compared with non-TN tumors and their functional and molecular interactions. Sixteen upregulated and 58 downregulated genes with a log fold change higher or equal to | 2 | were identified, including nine transcription factors. Coexpression network revealed EN1 as a hub gene, moreover Kaplan-Meier plotter survival analysis indicated that it was an appropriate prognostic marker for TN patients with breast cancer. Functional annotation analysis of protein-protein interaction network showed FOXM1 as an upexpressed and ESR1 as a downexpressed hub genes are suitable targets as far as antitumor protein therapy is concerned in TN breast cancers. The consensus analysis of two microRNA datasets revealed seven DE miRNAs. The gene-transcriptional factor (TF)-miRNA network revealed mir-135b and mir-29b are the hub nodes and involved in feedback loops with GATA3. This study suggests that dysregulated TFs and miRNAs have pivotal roles in regulation of TN oncotranscriptomic profile and might become both biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Farzaneh Darbeheshti
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Breast Cancer Association (BrCA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Disease Research Center, Fasa University of Medical Sciences, Fasa, Iran.,Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Javad Tavakkoly Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Ruan XH, Liu XM, Yang ZX, Zhang SP, Li QZ, Lin CS. INPP4B promotes colorectal cancer cell proliferation by activating mTORC1 signaling and cap-dependent translation. Onco Targets Ther 2019; 12:3109-3117. [PMID: 31114251 PMCID: PMC6485035 DOI: 10.2147/ott.s186365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background and objective Inositol polyphosphate 4-phosphatase type II (INPP4B) is over-expressed in CRC tissues, and emerges as an oncogene. However, the mechanism by which INPP4B regulates CRC cell proliferation remains largely unclear. In this study, we aimed to investigate the regulatory mechanisms of INPP4B in CRC. Materials and methods The expression levels of mRNA were detected by qRT-PCR. The expression levels of protein were determined by Western blot. Cell Counting Kit-8 (CCK-8) assays and BrdU incorporation assays were performed to evaluate cell proliferation abilities. Bicistronic luciferase assays and the m7GTP pull down assay were performed to measure the cap-dependent translation in cells. Results INPP4B promotes CRC cell proliferation by increasing mTORC1 activity. Furthermore, it was shown that the activation of mTORC1 signaling by INPP4B led to increased cap-dependent translation, which is essential for INPP4B-mediated CRC cell proliferation. Finally, it was demonstrated that increased AKT and serum and glucocorticoid-inducible kinase 1 activity contributed to the activation of cap-dependent translation induced by INPP4B. Conclusion Collectively, the present study reveals INPP4B promotes colorectal cancer cell proliferation by activating mTORC1 signaling and cap-dependent translation.
Collapse
Affiliation(s)
- Xin-Hua Ruan
- Department of Cardiac Surgery, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| | - Xi-Mei Liu
- Department of Cardiac Surgery, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| | - Zhi-Xiang Yang
- Department of Cardiac Surgery, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| | - Shao-Peng Zhang
- Department of Cardiac Surgery, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| | - Quan-Zheng Li
- Department of Cardiac Surgery, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| | - Chun-Sheng Lin
- Department of Medical Service, TianJin Union Medical Centre, Tianjin, People's Republic of China,
| |
Collapse
|
13
|
Asleh K, Lyck Carstensen S, Tykjaer Jørgensen CL, Burugu S, Gao D, Won JR, Jensen MB, Balslev E, Laenkholm AV, Nielsen DL, Ejlertsen B, Nielsen TO. Basal biomarkers nestin and INPP4B predict gemcitabine benefit in metastatic breast cancer: Samples from the phase III SBG0102 clinical trial. Int J Cancer 2018; 144:2578-2586. [PMID: 30411790 DOI: 10.1002/ijc.31969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/04/2018] [Accepted: 10/23/2018] [Indexed: 01/14/2023]
Abstract
In a formal prospective-retrospective analysis of the phase III SBG0102 clinical trial randomizing metastatic breast cancer patients to gemcitabine-docetaxel or to single agent docetaxel, patients with basal-like tumors by PAM50 gene expression had significantly better overall survival in the gemcitabine arm. By immunohistochemistry (IHC), triple negative status was not predictive, but more specific biomarkers have since become available defining basal-like by nestin positivity or loss of inositol-polyphosphate-4-phosphate (INPP4B). Here, we evaluate their capacity to identify which patients benefit from gemcitabine in the metastatic setting. Nestin and INPP4B staining and interpretation followed published methods. A prespecified statistical plan evaluated the primary hypothesis that patients with basal-like breast cancer, defined as "nestin+ or INPP4B-", would have superior overall survival on gemcitabine-docetaxel when compared to docetaxel. Interaction tests, Kaplan-Meier curves and forest plots were used to assess prognostic and predictive capacities of biomarkers relative to treatment. Among 239 cases evaluable for our study, 36 (15%) had been classified as basal-like by PAM50. "Nestin+ or INPP4B-" was observed in 41 (17%) of the total cases and was significantly associated with PAM50 basal-like subtype. Within an estimated median follow-up of 13 years, patients assigned as IHC basal "nestin+ or INPP4B-" had significantly better overall survival on gemcitabine-docetaxel versus docetaxel monotherapy (HR = 0.31, 95%CI: 0.16-0.60), whereas no differences were observed for other patients (HR = 0.99), p-interaction < 0.01. In the metastatic setting, women with IHC basal breast cancers defined as "nestin+ or INPP4B-" have superior overall survival when randomized to gemcitabine-containing chemotherapy compared to docetaxel alone. These findings need to be validated using larger prospective-retrospective phase III clinical trials series.
Collapse
Affiliation(s)
- Karama Asleh
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada
| | | | | | - Samantha Burugu
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada
| | - Dongxia Gao
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada
| | - Jennifer R Won
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada.,Canadian Immunohistochemistry Quality Control, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Eva Balslev
- Department of Pathology, Herlev and Gentofte Hospital, Herlev, Denmark
| | | | - Dorte L Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Cooperative Group, Copenhagen, Denmark
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada
| |
Collapse
|
14
|
Tan P, Ye Y, He L, Xie J, Jing J, Ma G, Pan H, Han L, Han W, Zhou Y. TRIM59 promotes breast cancer motility by suppressing p62-selective autophagic degradation of PDCD10. PLoS Biol 2018; 16:e3000051. [PMID: 30408026 PMCID: PMC6245796 DOI: 10.1371/journal.pbio.3000051] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/20/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer cells adopt various modes of migration during metastasis. How the ubiquitination machinery contributes to cancer cell motility remains underexplored. Here, we report that tripartite motif (TRIM) 59 is frequently up-regulated in metastatic breast cancer, which is correlated with advanced clinical stages and reduced survival among breast cancer patients. TRIM59 knockdown (KD) promoted apoptosis and inhibited tumor growth, while TRIM59 overexpression led to the opposite effects. Importantly, we uncovered TRIM59 as a key regulator of cell contractility and adhesion to control the plasticity of metastatic tumor cells. At the molecular level, we identified programmed cell death protein 10 (PDCD10) as a target of TRIM59. TRIM59 stabilized PDCD10 by suppressing RING finger and transmembrane domain-containing protein 1 (RNFT1)-induced lysine 63 (K63) ubiquitination and subsequent phosphotyrosine-independent ligand for the Lck SH2 domain of 62 kDa (p62)-selective autophagic degradation. TRIM59 promoted PDCD10-mediated suppression of Ras homolog family member A (RhoA)-Rho-associated coiled-coil kinase (ROCK) 1 signaling to control the transition between amoeboid and mesenchymal invasiveness. PDCD10 overexpression or administration of a ROCK inhibitor reversed TRIM59 loss-induced contractile phenotypes, thereby accelerating cell migration, invasion, and tumor formation. These findings establish the rationale for targeting deregulated TRIM59/PDCD10 to treat breast cancer.
Collapse
Affiliation(s)
- Peng Tan
- Department of Medical Oncology and Biomedical Research Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, United States of America
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, United States of America
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, United States of America
| | - Jiansheng Xie
- Department of Medical Oncology and Biomedical Research Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ji Jing
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, United States of America
| | - Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, United States of America
| | - Hongming Pan
- Department of Medical Oncology and Biomedical Research Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, United States of America
| | - Weidong Han
- Department of Medical Oncology and Biomedical Research Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, United States of America
- Department of Medical Physiology, College of Medicine, Texas A&M University, Temple, Texas, United States of America
| |
Collapse
|
15
|
Robert M, Frenel JS, Bourbouloux E, Berton Rigaud D, Patsouris A, Augereau P, Gourmelon C, Campone M. Efficacy of buparlisib in treating breast cancer. Expert Opin Pharmacother 2017; 18:2007-2016. [PMID: 29169282 DOI: 10.1080/14656566.2017.1410139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Breast cancer is the most frequent cancer in women. Despite a decline in breast cancer mortality, prognosis of advanced breast cancer remains poor. In a desperate need to improve breast cancer outcomes, newer agents that target molecular pathways are being tested. Deregulation of the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) is frequently found in breast cancer. This can lead to resistance of endocrine therapy and anti-HER2 therapies. Targeting this pathway may restore sensitivity to these compounds. Buparlisib (BKM-120) is an orally active pan-PI3K inhibitor evaluated in different tumor types. Areas covered: Buparlisib is one of the most investigated PI3K inhibitors. Preclinical and clinical studies of buparlisib in breast cancer are analyzed and discussed. This article reviews the status of buparlisib, completed and ongoing trials, and its safety. Expert opinion: PI3K inhibitors show promising results in breast cancer. However, we raise a number of issues including the identification of biomarkers to predict treatment response and strategies to counteract resistance. Moreover, its toxicity profile could limit its extensive use.
Collapse
Affiliation(s)
- Marie Robert
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Jean-Sébastien Frenel
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Emmanuelle Bourbouloux
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Dominique Berton Rigaud
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Anne Patsouris
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Paule Augereau
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Carole Gourmelon
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Mario Campone
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France.,c Nantes and Angers , Centre de Recherche en Cancérologie Nantes-Angers (CRNA) , France
| |
Collapse
|
16
|
Tessier-Cloutier B, Asleh-Aburaya K, Shah V, McCluggage WG, Tinker A, Gilks CB. Molecular subtyping of mammary-like adenocarcinoma of the vulva shows molecular similarity to breast carcinomas. Histopathology 2017; 71:446-452. [DOI: 10.1111/his.13239] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Basile Tessier-Cloutier
- Division of Anatomical Pathology; Vancouver General Hospital; University of British Columbia; Vancouver Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver Canada
| | - Karama Asleh-Aburaya
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver Canada
- Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver Canada
| | - Varsha Shah
- Department of Pathology; Royal Gwent Hospital; Newport UK
| | - W Glenn McCluggage
- Department of Pathology; Belfast Health and Social Care Trust; Belfast UK
| | - Anna Tinker
- Division of Medical Oncology; British Columbia Cancer Agency; Vancouver Canada
| | - C Blake Gilks
- Division of Anatomical Pathology; Vancouver General Hospital; University of British Columbia; Vancouver Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver Canada
| |
Collapse
|
17
|
Strotbek M, Schmid S, Sánchez-González I, Boerries M, Busch H, Olayioye MA. miR-181 elevates Akt signaling by co-targeting PHLPP2 and INPP4B phosphatases in luminal breast cancer. Int J Cancer 2017; 140:2310-2320. [DOI: 10.1002/ijc.30661] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/01/2017] [Accepted: 02/13/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Michaela Strotbek
- Institute of Cell Biology and Immunology, University of Stuttgart; 70569 Stuttgart Germany
| | - Simone Schmid
- Institute of Cell Biology and Immunology, University of Stuttgart; 70569 Stuttgart Germany
| | | | - Melanie Boerries
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg; 79104 Freiburg Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ); Heidelberg 69120 Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg; 79104 Freiburg Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ); Heidelberg 69120 Germany
- Lübeck Institute of Experimental Dermatology, University of Lübeck; Lübeck Germany
| | - Monilola A. Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart; 70569 Stuttgart Germany
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart; Stuttgart Germany
| |
Collapse
|
18
|
Xie BH, He X, Hua RX, Zhang B, Tan GS, Xiong SQ, Liu LS, Chen W, Yang JY, Wang XN, Li HP. Mir-765 promotes cell proliferation by downregulating INPP4B expression in human hepatocellular carcinoma. Cancer Biomark 2016; 16:405-13. [PMID: 27062697 DOI: 10.3233/cbm-160579] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
microRNAs (miRNAs) dysregulation is widely involved in cancer progression and contributed to sustained cell proliferation by directly targeting multiple targets. Therefore, better understanding the underlying mechanism of miRNA in carcinogenesis may improve diagnostic and therapeutic strategies for malignancy. In our study, we found that mir-765 is upregulated in both hepatocellular carcinoma (HCC) cell lines and tissues, compared to human normal liver cell line and adjacent non-cancerous tissues, respectively. Overexpression of mir-765 increased HCC cells proliferation and tumorigenicity, whereas inhibition of mir-765 reverses this effect. Furthermore, we demonstrated that INPP4B as a direct target of mir-765 and ectopic expression of mir-765 repressed INPP4B expression, resulting in upregulation of p-AKT, Cyclin D1, and downregulation of p-FOXO3a, p21 expression in HCC. Strikingly, we found that silencing the expression of INPP4B is the essential biological function of miR-765 during HCC cell proliferation. Collectively, our findings reveal that miR-765 is a potential onco-miR that participates in carcinogenesis of human HCC by suppressing INPP4B expression, and might represent a potential therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Bin-Hui Xie
- Department of General Surgery, the First Affiliated Hospital of Gannan Medical University, Guangzhou, Guangdong, China
| | - Xiao He
- Department of General Surgery, the First Affiliated Hospital of Gannan Medical University, Guangzhou, Guangdong, China
| | - Rui-Xi Hua
- Department of Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bing Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Guo-Sheng Tan
- Department of Interventional Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shi-Qiu Xiong
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Liang-Shuai Liu
- Department of Interventional Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wei Chen
- Department of Interventional Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Yong Yang
- Department of Interventional Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Nong Wang
- Department of General Surgery, the First Affiliated Hospital of Gannan Medical University, Guangzhou, Guangdong, China
| | - He-Ping Li
- Department of Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,Department of Interventional Radiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Kwon J, Kim DH, Park JM, Park YH, Hwang YH, Wu HG, Shin KH, Kim IA. Targeting Phosphatidylinositol 4-Kinase IIIα for Radiosensitization: A Potential Model of Drug Repositioning Using an Anti-Hepatitis C Viral Agent. Int J Radiat Oncol Biol Phys 2016; 96:867-876. [DOI: 10.1016/j.ijrobp.2016.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022]
|
20
|
Emerging evidence of signalling roles for PI(3,4)P2 in Class I and II PI3K-regulated pathways. Biochem Soc Trans 2016; 44:307-14. [PMID: 26862220 DOI: 10.1042/bst20150248] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There are eight members of the phosphoinositide family of phospholipids in eukaryotes; PI, PI3P, PI4P, PI5P, PI(4,5)P2, PI(3,4)P2, PI(3,5)P2 and PI(3,4,5)P3. Receptor activation of Class I PI3Ks stimulates the phosphorylation of PI(4,5)P2 to form PI(3,4,5)P3. PI(3,4,5)P3 is an important messenger molecule that is part of a complex signalling network controlling cell growth and division. PI(3,4,5)P3 can be dephosphorylated by both 3- and 5-phosphatases, producing PI(4,5)P2 and PI(3,4)P2, respectively. There is now strong evidence that PI(3,4)P2 generated by this route does not merely represent another pathway for removal of PI(3,4,5)P3, but can act as a signalling molecule in its own right, regulating macropinocytosis, fast endophilin-mediated endocytosis (FEME), membrane ruffling, lamellipodia and invadopodia. PI(3,4)P2 can also be synthesized directly from PI4P by Class II PI3Ks and this is important for the maturation of clathrin-coated pits [clathrin-mediated endocytosis (CME)] and signalling in early endosomes. Thus PI(3,4)P2 is emerging as an important signalling molecule involved in the coordination of several specific membrane and cytoskeletal responses. Further, its inappropriate accumulation contributes to pathology caused by mutations in genes encoding enzymes responsible for its degradation, e.g. Inpp4B.
Collapse
|
21
|
Asleh-Aburaya K, Sheffield BS, Kos Z, Won JR, Wang XQ, Gao D, Wolber R, Gilks CB, Bernard PS, Chia SKL, Nielsen TO. Basal biomarkers nestin and INPP4b identify intrinsic subtypes accurately in breast cancers that are weakly positive for oestrogen receptor. Histopathology 2016; 70:185-194. [DOI: 10.1111/his.13038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/10/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Karama Asleh-Aburaya
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
| | - Brandon S Sheffield
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
| | - Zuzana Kos
- Department of Pathology and Laboratory Medicine; University of Ottawa and The Ottawa Hospital; Ottawa ON Canada
| | - Jennifer R Won
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
- Department of Pathology and Laboratory Medicine; Canadian Immunohistochemistry Quality Control (CIQC); University of British Columbia; Vancouver BC Canada
| | - Xiu Q Wang
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
| | - Dongxia Gao
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
| | - Robert Wolber
- Department of Pathology and Laboratory Medicine; Lions Gate Hospital; North Vancouver BC Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
- Department of Pathology and Laboratory Medicine; Canadian Immunohistochemistry Quality Control (CIQC); University of British Columbia; Vancouver BC Canada
| | - Philip S Bernard
- Department of Pathology; University of Utah/Health Sciences; Salt Lake City UT USA
| | - Stephen K L Chia
- Department of Medical Oncology; British Columbia Cancer Agency; Vancouver BC Canada
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine; Genetic Pathology Evaluation Centre; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
22
|
Wang P, Ma D, Wang J, Fang Q, Gao R, Wu W, Cao L, Hu X, Zhao J, Li Y. INPP4B-mediated DNA repair pathway confers resistance to chemotherapy in acute myeloid leukemia. Tumour Biol 2016; 37:12513-12523. [PMID: 27342972 DOI: 10.1007/s13277-016-5111-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/09/2016] [Indexed: 10/21/2022] Open
Abstract
INPP4B has been recently shown to be a poor prognostic marker and confer chemo- or radio-resistance in AML cells, whereas, the underlying mechanisms remain unclear. Herein, we aimed to explore the possible mechanisms mediated the resistance to chemotherapy in AML. We found that INPP4B-mediated resistance to genotoxic drug, cytarabine, was accompanied by lower p-H2AX accumulation in KG-1 cells, and INPP4B knockdown evidently sensitized KG-1 cells to cytarabine, meanwhile, p-H2AX expression was increased dramatically. Then, we observed that INPP4B knockdown inhibited the loss of p-H2AX expression after cytarabine removal in INPP4B-silenced KG-1 cells, whereas, in control KG-1 cells, the expression of p-H2AX was reduced in a time-dependent manner. Next, INPP4B knockdown can significantly downregulate ATM expression and subsequently inhibit the activation of ATM downstream targets of p-ATM, p-BRCA1, p-ATR, and p-RAD51. Furthermore, nuclear localization of p65 was inhibited after INPP4B knockdown, and reactivation of p65 can rescue the INPP4B knockdown-induced inhibition of ATM, p-ATM, p-BRCA1, p-ATR, and p-RAD51. Finally, INPP4B expression was positively correlated with ATM expression in AML cells, both INPP4B knockdown and KU55933 can significantly sensitize primary myeloid leukemic cells to cytarabine treatment.Collectively, these data suggest that enhanced ATM-dependent DNA repair is involved in resistance to chemotherapy in INPP4Bhigh AML, which could be mediated by p65 nuclear translocation, combination chemotherapy with INPP4B or DNA repair pathway inhibition represents a promising strategy in INPP4Bhigh AML.
Collapse
Affiliation(s)
- Ping Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,Department of Pharmacy, Affiliated BaiYun Hospital of Guizhou Medical University, Guiyang, 550014, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China. .,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China. .,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Qin Fang
- Department of Pharmacy, Affiliated BaiYun Hospital of Guizhou Medical University, Guiyang, 550014, China.,Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Rui Gao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Weibing Wu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Lu Cao
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Xiuying Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jiangyuan Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yan Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Key Laboratory of Hematological Disease Diagnostic and Treat Centre of GuiZhou Province, Guiyang, 550004, China.,GuiZhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
23
|
Chen H, Li H, Chen Q. INPP4B reverses docetaxel resistance and epithelial-to-mesenchymal transition via the PI3K/Akt signaling pathway in prostate cancer. Biochem Biophys Res Commun 2016; 477:467-72. [PMID: 27318090 DOI: 10.1016/j.bbrc.2016.06.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
Docetaxel efficiency in the therapy of prostate cancer (PCa) patients is limited due to the development of chemoresistance. Recent studies have implied a role of INPP4B in tumor chemoresistance, while the effects of INPP4B on docetaxel resistance in PCa have not been elucidated. In the present study, the docetaxel-resistant human PCa cell lines PC3-DR and DU-145-DR were established from the parental cell lines PC3 and DU-145, and the expression and role of INPP4B in docetaxel-resistant PCa cells were investigated. The results demonstrated that INPP4B expression was significantly downregulated in docetaxel-resistant cells. Overexpression of INPP4B increased the sensitivity to docetaxel and promoted cell apoptosis in PC3-DR and DU-145-DR cells. In addition, INPP4B overexpression downregulated the expression of the mesenchymal markers fibronectin, N-cadherin, and vimentin, and upregulated the expression level of the epithelial maker E-cadherin. Furthermore, INPP4B overexpression markedly inhibited the PI3K/Akt pathway. We also found that IGF-1, the inhibitor of PI3K/Akt, markedly blocked the change in EMT markers induced by overexpression of INPP4B, and reversed the resistance of PC3-DR and DU-145-DR cells to docetaxel, which is sensitized by Flag-INPP4B. In summary, the presented data indicate that INPP4B is crucial for docetaxel-resistant PCa cell survival, potentially by regulating EMT through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Haiwen Chen
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiao Tong University, PR China
| | - Hongliang Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiao Tong University, PR China.
| | - Qi Chen
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiao Tong University, PR China
| |
Collapse
|
24
|
Hu QL, Wang HB, Yang M. Significance of expression of INPP4B in gastric cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:2478-2484. [DOI: 10.11569/wcjd.v24.i16.2478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of inositol polyphosphate-4-phosphatase, type II (INPP4B) in gastric cancer, and to analyze its relationship with clinical and pathological characteristics.
METHODS: The expression of INPP4B mRNA and protein in 50 gastric cancer tissues and matched tumor-adjacent normal tissues was detected by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC), respectively. The relationship between INPP4B expression and clinical and pathological characteristics was then analyzed.
RESULTS: INPP4B mRNA expression was significantly lower in gastric cancer tissue than in adjacent normal tissues (P < 0.01). The expression of INPP4B protein in gastric cancer tissues was also significantly lower compared with adjacent normal tissues (28.0% vs 82.0%, P < 0.01). The expression of INPP4B mRNA and protein was significantly related to tumor differentiation, lymph node metastasis and TNM stage in GC (P < 0.05), but not to gender, age or tumor size (P > 0.05).
CONCLUSION: Both INPP4B protein and mRNA are down-regulated in gastric cancer, and its expression significantly correlates with tumor differentiation, lymph node metastasis and TNM stage. INPP4B may be a tumor suppressor gene for gastric cancer.
Collapse
|
25
|
Ding X, Faber K, Shi Y, McKnight J, Dorshorst D, Ware JA, Dean B. Validation and determination of taselisib, a β-sparing phosphoinositide 3-kinase (PI3K) inhibitor, in human plasma by LC-MS/MS. J Pharm Biomed Anal 2016; 126:117-23. [PMID: 27187764 DOI: 10.1016/j.jpba.2016.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/13/2016] [Accepted: 04/19/2016] [Indexed: 11/29/2022]
Abstract
A liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method for the determination of taselisib (GDC-0032, RO5537381) concentrations in human plasma has been developed and validated to support bioanalysis of clinical samples. Solid phase extraction (SPE) was used to extract plasma samples (50μL) and the resulting samples were analyzed using reversed phase chromatography and mass spectrometry coupled with an atmospheric pressure chemical ionization interface. The mass analysis of taselisib was performed using multiple reaction monitoring transitions in positive ionization mode. The method was validated over the calibration curve range 0.400-400ng/mL using linear regression and 1/x(2) weighting. The within-run relative standard deviation (%RSD) ranged from 1.3 to 5.6%, while the between-run %RSD varied from 2.0 to 4.5% for LLOQ, low, medium, medium high and high QCs. The accuracy ranged from 94.7 to 100.3% of nominal for within-run and 96.0-99.0% of nominal for between-run for the same QCs. Extraction recovery of taselisib was between 83.8% and 92.9%. Stability of taselisib was established in human plasma for 977days at -20°C and -70°C and established in sample extracts for 96h when stored at 2 - 8°C. Stable-labeled internal standard was used to minimize matrix effects. Mean single dose pharmacokinetic parameters determined using this method for a phase I/II clinical trial were: Cmax=35.2ng/mL, AUC0-inf=1570ngh/mL, and T1/2=39.3h.
Collapse
Affiliation(s)
- X Ding
- Genentech, Drug Metabolism and Pharmacokinetics, 1 DNA Way, South San Francisco, CA 94080, United States.
| | - K Faber
- Genentech, Clinical Pharmacology, 1 DNA Way, South San Francisco, CA 94080, United States
| | - Y Shi
- Covance Laboratories, 3301 Kinsman Blvd., Madison, WI 53704, United States
| | - J McKnight
- Covance Laboratories, 3301 Kinsman Blvd., Madison, WI 53704, United States
| | - D Dorshorst
- Covance Laboratories, 3301 Kinsman Blvd., Madison, WI 53704, United States
| | - J A Ware
- Genentech, Clinical Pharmacology, 1 DNA Way, South San Francisco, CA 94080, United States
| | - B Dean
- Genentech, Drug Metabolism and Pharmacokinetics, 1 DNA Way, South San Francisco, CA 94080, United States
| |
Collapse
|
26
|
den Hollander P, Rawls K, Tsimelzon A, Shepherd J, Mazumdar A, Hill J, Fuqua SAW, Chang JC, Osborne CK, Hilsenbeck SG, Mills GB, Brown PH. Phosphatase PTP4A3 Promotes Triple-Negative Breast Cancer Growth and Predicts Poor Patient Survival. Cancer Res 2016; 76:1942-53. [PMID: 26921331 PMCID: PMC4873402 DOI: 10.1158/0008-5472.can-14-0673] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/15/2015] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) has the worst prognosis of all breast cancers, and women diagnosed with TNBC currently lack targeted treatment options. To identify novel targets for TNBC, we evaluated phosphatase expression in breast tumors and characterized their contributions to in vitro and in vivo growth of TNBC. Using Affymetrix microarray analysis of 102 breast cancers, we identified 146 phosphatases that were significantly differentially expressed in TNBC compared with estrogen receptor (ER)-positive tumors. Of these, 19 phosphatases were upregulated (0.66-fold; FDR = 0.05) in TNBC compared with ER-positive breast cancers. We knocked down 17 overexpressed phosphatases in four triple-negative and four ER-positive breast cancer lines using specific siRNAs and found that depletion of six of these phosphatases significantly reduced growth and anchorage-independent growth of TNBC cells to a greater extent than ER-positive cell lines. Further analysis of the phosphatase PTP4A3 (also known as PRL-3) demonstrated its requirement for G1-S cell-cycle progression in all breast cancer cells, but PTP4A3 regulated apoptosis selectively in TNBC cells. In addition, PTP4A3 inhibition reduced the growth of TNBC tumors in vivo Moreover, in silico analysis revealed the PTP4A3 gene to be amplified in 29% of basal-like breast cancers, and high expression of PTP4A3 could serve as an independent prognostic indicator for worse overall survival. Collectively, these studies define the importance of phosphatase overexpression in TNBC and lay the foundation for the development of new targeted therapies directed against phosphatases or their respective signaling pathways for TNBC patients. Cancer Res; 76(7); 1942-53. ©2016 AACR.
Collapse
Affiliation(s)
- Petra den Hollander
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn Rawls
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jonathan Shepherd
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jenny C Chang
- Methodist Cancer Center, The Methodist Hospital Research Institute, Houston, Texas
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
27
|
Josephs DH, Sarker D. Pharmacodynamic Biomarker Development for PI3K Pathway Therapeutics. TRANSLATIONAL ONCOGENOMICS 2016; 7:33-49. [PMID: 26917948 PMCID: PMC4762492 DOI: 10.4137/tog.s30529] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway is integral to many essential cell processes, including cell growth, differentiation, proliferation, motility, and metabolism. Somatic mutations and genetic amplifications that result in activation of the pathway are frequently detected in cancer. This has led to the development of rationally designed therapeutics targeting key members of the pathway. Critical to the successful development of these drugs are pharmacodynamic biomarkers that aim to define the degree of target and pathway inhibition. In this review, we discuss the pharmacodynamic biomarkers that have been utilized in early-phase clinical trials of PI3K pathway inhibitors. We focus on the challenges related to development and interpretation of these assays, their optimal integration with pharmacokinetic and predictive biomarkers, and future strategies to ensure successful development of PI3K pathway inhibitors within a personalized medicine paradigm for cancer.
Collapse
Affiliation(s)
- Debra H Josephs
- Department of Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | - Debashis Sarker
- Department of Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
28
|
Biological and clinical significance of loss of heterozygosity at the INPP4B gene locus in Japanese breast cancer. Breast 2016; 25:62-8. [DOI: 10.1016/j.breast.2015.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/07/2015] [Accepted: 10/20/2015] [Indexed: 01/21/2023] Open
|
29
|
Liu X, Cohen JI. The role of PI3K/Akt in human herpesvirus infection: From the bench to the bedside. Virology 2015; 479-480:568-77. [PMID: 25798530 PMCID: PMC4424147 DOI: 10.1016/j.virol.2015.02.040] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 12/25/2022]
Abstract
The phosphatidylinositol-3-kinase (PI3K)-Akt signaling pathway regulates several key cellular functions including protein synthesis, cell growth, glucose metabolism, and inflammation. Many viruses have evolved mechanisms to manipulate this signaling pathway to ensure successful virus replication. The human herpesviruses undergo both latent and lytic infection, but differ in cell tropism, growth kinetics, and disease manifestations. Herpesviruses express multiple proteins that target the PI3K/Akt cell signaling pathway during the course of their life cycle to facilitate viral infection, replication, latency, and reactivation. Rare human genetic disorders with mutations in either the catalytic or regulatory subunit of PI3K that result in constitutive activation of the protein predispose to severe herpesvirus infections as well as to virus-associated malignancies. Inhibiting the PI3K/Akt pathway or its downstream proteins using drugs already approved for other diseases can block herpesvirus lytic infection and may reduce malignancies associated with latent herpesvirus infections.
Collapse
Affiliation(s)
- XueQiao Liu
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeffrey I Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
30
|
Mendelian disorders of PI metabolizing enzymes. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:867-81. [PMID: 25510381 DOI: 10.1016/j.bbalip.2014.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/18/2014] [Accepted: 12/01/2014] [Indexed: 12/18/2022]
Abstract
More than twenty different genetic diseases have been described that are caused by mutations in phosphoinositide metabolizing enzymes, mostly in phosphoinositide phosphatases. Although generally ubiquitously expressed, mutations in these enzymes, which are mainly loss-of-function, result in tissue-restricted clinical manifestations through mechanisms that are not completely understood. Here we analyze selected disorders of phosphoinositide metabolism grouped according to the principle tissue affected: the nervous system, muscle, kidney, the osteoskeletal system, the eye, and the immune system. We will highlight what has been learnt so far from the study of these disorders about not only the cellular and molecular pathways that are involved or are governed by phosphoinositides, but also the many gaps that remain to be filled to gain a full understanding of the pathophysiological mechanisms underlying the clinical manifestations of this steadily growing class of diseases, most of which still remain orphan in terms of treatment. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
|
31
|
Pelicano H, Zhang W, Liu J, Hammoudi N, Dai J, Xu RH, Pusztai L, Huang P. Mitochondrial dysfunction in some triple-negative breast cancer cell lines: role of mTOR pathway and therapeutic potential. Breast Cancer Res 2014; 16:434. [PMID: 25209360 PMCID: PMC4303115 DOI: 10.1186/s13058-014-0434-6] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 08/27/2014] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is a subtype of highly malignant breast cancer with poor prognosis. TNBC is not amenable to endocrine therapy and often exhibit resistance to current chemotherapeutic agents, therefore, further understanding of the biological properties of these cancer cells and development of effective therapeutic approaches are urgently needed. METHODS We first investigated the metabolic alterations in TNBC cells in comparison with other subtypes of breast cancer cells using molecular and metabolic analyses. We further demonstrated that targeting these alterations using specific inhibitors and siRNA approach could render TNBC cells more sensitive to cell death compared to other breast cancer subtypes. RESULTS We found that TNBC cells compared to estrogen receptor (ER) positive cells possess special metabolic characteristics manifested by high glucose uptake, increased lactate production, and low mitochondrial respiration which is correlated with attenuation of mTOR pathway and decreased expression of p70S6K. Re-expression of p70S6K in TNBC cells reverses their glycolytic phenotype to an active oxidative phosphorylation (OXPHOS) state, while knockdown of p70S6K in ER positive cells leads to suppression of mitochondrial OXPHOS. Furthermore, lower OXPHOS activity in TNBC cells renders them highly dependent on glycolysis and the inhibition of glycolysis is highly effective in targeting TNBC cells despite their resistance to other anticancer agents. CONCLUSIONS Our study shows that TNBC cells have profound metabolic alterations characterized by decreased mitochondrial respiration and increased glycolysis. Due to their impaired mitochondrial function, TNBC cells are highly sensitive to glycolytic inhibition, suggesting that such metabolic intervention may be an effective therapeutic strategy for this subtype of breast cancer cells.
Collapse
|
32
|
Abstract
The specific interaction of phosphoinositides with proteins is critical for a plethora of cellular processes, including cytoskeleton remodelling, mitogenic signalling, ion channel regulation and membrane traffic. The spatiotemporal restriction of different phosphoinositide species helps to define compartments within the cell, and this is particularly important for membrane trafficking within both the secretory and endocytic pathways. Phosphoinositide homoeostasis is tightly regulated by a large number of inositol kinases and phosphatases, which respectively phosphorylate and dephosphorylate distinct phosphoinositide species. Many of these enzymes have been implicated in regulating membrane trafficking and, accordingly, their dysregulation has been linked to a number of human diseases. In the present review, we focus on the inositol phosphatases, concentrating on their roles in membrane trafficking and the human diseases with which they have been associated.
Collapse
|
33
|
Yuen JWF, Chung GTY, Lun SWM, Cheung CCM, To KF, Lo KW. Epigenetic inactivation of inositol polyphosphate 4-phosphatase B (INPP4B), a regulator of PI3K/AKT signaling pathway in EBV-associated nasopharyngeal carcinoma. PLoS One 2014; 9:e105163. [PMID: 25126743 PMCID: PMC4134277 DOI: 10.1371/journal.pone.0105163] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/21/2014] [Indexed: 12/23/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common viral-associated neoplasm in which multiple signaling cascades are interfered with by Epstein-Bar virus (EBV) latent proteins and various genetic alterations. Aside from the previously reported PIK3CA amplification, we examined the role of INPP4B, a negative regulator of the PI3K/AKT signaling pathway in the development of NPC. By RT-PCR and Western blotting, we revealed that the expression of INPP4B was down-regulated in all five established EBV-positive tumor lines. While INPP4B was consistently expressed in normal nasopharyngeal epithelial cells, downregulation of INPP4B was found in 32/65 (49.2%) of primary tumors by immunohistochemistry. Furthermore, our study also demonstrated the hypermethylation of the 5'CpG island of INPP4B in the tumors in which INPP4B transcription was downregulated. Notably, the re-expression of INPP4B was detected in the NPC cells treated with the demethylation agent (5-aza-2'deoxycytidine). Our study showed that promoter hypermethylation was the major mechanism for transcriptional silencing of INPP4B in NPC. Furthermore, restoration of INPP4B expression significantly suppressed PI3K/AKT downstream signals in the NPC C666-1 cells. In vivo growth inhibition was clearly demonstrated in the tumor cells stably expressing INPP4B. The findings indicate that epigenetic inactivation of INPP4B is one of the key mechanisms in activating PI3K/AKT signaling cascade and playing a role in the tumorigenesis of NPC.
Collapse
Affiliation(s)
- Jessie Wai-Fong Yuen
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
| | - Grace Tin-Yun Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
| | - Samantha Wei-Man Lun
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
| | - Chartia Ching-Mei Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
- Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong SAR
| | - Kwok-Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong SAR
- Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong SAR
- * E-mail:
| |
Collapse
|
34
|
Paplomata E, O'Regan R. The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers. Ther Adv Med Oncol 2014; 6:154-66. [PMID: 25057302 DOI: 10.1177/1758834014530023] [Citation(s) in RCA: 373] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The phosphoinositide 3 kinase (PI3K)/Akt/mammalian (or mechanistic) target of rapamycin (mTOR) pathway is a complicated intracellular pathway, which leads to cell growth and tumor proliferation and plays a significant role in endocrine resistance in breast cancer. Multiple compounds targeting this pathway are being evaluated in clinical trials. These agents are generally well tolerated and can be used in combination with targeted therapies, endocrine therapy or cytotoxic agents. The identification of subtypes of tumors more likely to respond to these therapeutics cannot be overemphasized, since breast cancer is a very heterogeneous malignancy. Activation of pathways such as KRAS and MEK can act as escape mechanisms that lead to resistance, thus a combination of agents targeting multiple steps of the intracellular machinery is promising. There is evidence that tumors with PIK3CA mutations are more sensitive to inhibitors of the PI3K pathway but this has yet to be validated. Large clinical trials with correlative studies are necessary to identify reliable biomarkers of efficacy.
Collapse
Affiliation(s)
| | - Ruth O'Regan
- Winship Cancer Institute of Emory University, 1365C Clifton Road, Atlanta, GA 30329, USA
| |
Collapse
|
35
|
Russo J, Santucci-Pereira J, Russo IH. The genomic signature of breast cancer prevention. Genes (Basel) 2014; 5:65-83. [PMID: 24705287 PMCID: PMC3978512 DOI: 10.3390/genes5010065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/31/2014] [Accepted: 02/08/2014] [Indexed: 11/16/2022] Open
Abstract
The breast of parous postmenopausal women exhibits a specific signature that has been induced by a full term pregnancy. This signature is centered in chromatin remodeling and the epigenetic changes induced by methylation of specific genes which are important regulatory pathways induced by pregnancy. Through the analysis of the genes found to be differentially methylated between women of varying parity, multiple positions at which beta-catenin production and use is inhibited were recognized. The biological importance of the pathways identified in this specific population cannot be sufficiently emphasized because they could represent a safeguard mechanism mediating the protection of the breast conferred by full term pregnancy.
Collapse
Affiliation(s)
- Jose Russo
- The Irma H. Russo MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| | - Julia Santucci-Pereira
- The Irma H. Russo MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| | - Irma H Russo
- The Irma H. Russo MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
36
|
Huemer F, Bartsch R, Gnant M. The PI3K/AKT/MTOR Signaling Pathway: The Role of PI3K and AKT Inhibitors in Breast Cancer. CURRENT BREAST CANCER REPORTS 2014. [DOI: 10.1007/s12609-014-0139-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
37
|
Sun Y, Ding H, Liu X, Li X, Li L. INPP4B overexpression enhances the antitumor efficacy of PARP inhibitor AG014699 in MDA-MB-231 triple-negative breast cancer cells. Tumour Biol 2014; 35:4469-77. [DOI: 10.1007/s13277-013-1589-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022] Open
|
38
|
Perets R, Wyant GA, Muto KW, Bijron JG, Poole BB, Chin KT, Chen JYH, Ohman AW, Stepule CD, Kwak S, Karst AM, Hirsch MS, Setlur SR, Crum CP, Dinulescu DM, Drapkin R. Transformation of the fallopian tube secretory epithelium leads to high-grade serous ovarian cancer in Brca;Tp53;Pten models. Cancer Cell 2013; 24:751-65. [PMID: 24332043 PMCID: PMC3917315 DOI: 10.1016/j.ccr.2013.10.013] [Citation(s) in RCA: 438] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/17/2013] [Accepted: 10/22/2013] [Indexed: 02/08/2023]
Abstract
High-grade serous ovarian carcinoma presents significant clinical and therapeutic challenges. Although the traditional model of carcinogenesis has focused on the ovary as a tumor initiation site, recent studies suggest that there may be additional sites of origin outside the ovary, namely the secretory cells of the fallopian tube. Our study demonstrates that high-grade serous tumors can originate in fallopian tubal secretory epithelial cells and also establishes serous tubal intraepithelial carcinoma as the precursor lesion to high-grade serous ovarian and peritoneal carcinomas in animal models targeting the Brca, Tp53, and Pten genes. These findings offer an avenue to address clinically important questions that are critical for cancer prevention and early detection in women carrying BRCA1 and BRCA2 mutations.
Collapse
Affiliation(s)
- Ruth Perets
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Gregory A Wyant
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine W Muto
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan G Bijron
- Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Barish B Poole
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth T Chin
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Yun H Chen
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Anders W Ohman
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Corey D Stepule
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Soongu Kwak
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alison M Karst
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Michelle S Hirsch
- Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sunita R Setlur
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher P Crum
- Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela M Dinulescu
- Eugene Braunwald Research Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Ronny Drapkin
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Viernes DR, Choi LB, Kerr WG, Chisholm JD. Discovery and development of small molecule SHIP phosphatase modulators. Med Res Rev 2013; 34:795-824. [PMID: 24302498 DOI: 10.1002/med.21305] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Inositol phospholipids play an important role in the transfer of signaling information across the cell membrane in eukaryotes. These signals are often governed by the phosphorylation patterns on the inositols, which are mediated by a number of inositol kinases and phosphatases. The src homology 2 (SH2) containing inositol 5-phosphatase (SHIP) plays a central role in these processes, influencing signals delivered through the PI3K/Akt/mTOR pathway. SHIP modulation by small molecules has been implicated as a treatment in a number of human disease states, including cancer, inflammatory diseases, diabetes, atherosclerosis, and Alzheimer's disease. In addition, alteration of SHIP phosphatase activity may provide a means to facilitate bone marrow transplantation and increase blood cell production. This review discusses the cellular signaling pathways and protein-protein interactions that provide the molecular basis for targeting the SHIP enzyme in these disease states. In addition, a comprehensive survey of small molecule modulators of SHIP1 and SHIP2 is provided, with a focus on the structure, potency, selectivity, and solubility properties of these compounds.
Collapse
Affiliation(s)
- Dennis R Viernes
- Department of Chemistry, Syracuse University, Syracuse, NY, USA 13244
| | - Lydia B Choi
- Department of Chemistry, Syracuse University, Syracuse, NY, USA 13244
| | - William G Kerr
- Department of Chemistry, Syracuse University, Syracuse, NY, USA 13244.,Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA 13210.,Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA 13210
| | - John D Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY, USA 13244
| |
Collapse
|
40
|
Ikonomov OC, Filios C, Sbrissa D, Chen X, Shisheva A. The PIKfyve-ArPIKfyve-Sac3 triad in human breast cancer: Functional link between elevated Sac3 phosphatase and enhanced proliferation of triple negative cell lines. Biochem Biophys Res Commun 2013; 440:342-7. [PMID: 24070605 DOI: 10.1016/j.bbrc.2013.09.080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 09/16/2013] [Indexed: 12/11/2022]
Abstract
The phosphoinositide 5-kinase PIKfyve and 5-phosphatase Sac3 are scaffolded by ArPIKfyve in the PIKfyve-ArPIKfyve-Sac3 (PAS) regulatory complex to trigger a unique loop of PtdIns3P-PtdIns(3,5)P2 synthesis and turnover. Whereas the metabolizing enzymes of the other 3-phosphoinositides have already been implicated in breast cancer, the role of the PAS proteins and the PtdIns3P-PtdIns(3,5)P2 conversion is unknown. To begin elucidating their roles, in this study we monitored the endogenous levels of the PAS complex proteins in cell lines derived from hormone-receptor positive (MCF7 and T47D) or triple-negative breast cancers (TNBC) (BT20, BT549 and MDA-MB-231) as well as in MCF10A cells derived from non-tumorigenic mastectomy. We report profound upregulation of Sac3 and ArPIKfyve in the triple negative vs. hormone-sensitive breast cancer or non-tumorigenic cells, with BT cell lines showing the highest levels. siRNA-mediated knockdown of Sac3, but not that of PIKfyve, significantly inhibited proliferation of BT20 and BT549 cells. In these cells, knockdown of ArPIKfyve had only a minor effect, consistent with a primary role for Sac3 in TNBC cell proliferation. Intriguingly, steady-state levels of PtdIns(3,5)P2 in BT20 and T47D cells were similar despite the 6-fold difference in Sac3 levels between these cell lines. However, steady-state levels of PtdIns3P and PtdIns5P, both regulated by the PAS complex, were significantly reduced in BT20 vs. T47D or MCF10A cell lines, consistent with elevated Sac3 affecting directly or indirectly the homeostasis of these lipids in TNBC. Together, our results uncover an unexpected role for Sac3 phosphatase in TNBC cell proliferation. Database analyses, discussed herein, reinforce the involvement of Sac3 in breast cancer pathogenesis.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States.
| | | | | | | | | |
Collapse
|
41
|
Hoshiba T, Tanaka M. Breast cancer cell behaviors on staged tumorigenesis-mimicking matrices derived from tumor cells at various malignant stages. Biochem Biophys Res Commun 2013; 439:291-6. [PMID: 23978418 DOI: 10.1016/j.bbrc.2013.08.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 08/11/2013] [Indexed: 12/21/2022]
Abstract
Extracellular matrix (ECM) has been focused to understand tumor progression in addition to the genetic mutation of cancer cells. Here, we prepared "staged tumorigenesis-mimicking matrices" which mimic in vivo ECM in tumor tissue at each malignant stage to understand the roles of ECM in tumor progression. Breast tumor cells, MDA-MB-231 (invasive), MCF-7 (non-invasive), and MCF-10A (benign) cells, were cultured to form their own ECM beneath the cells and formed ECM was prepared as staged tumorigenesis-mimicking matrices by decellularization treatment. Cells showed weak attachment on the matrices derived from MDA-MB-231 cancer cells. The proliferations of MDA-MB-231 and MCF-7 was promoted on the matrices derived from MDA-MB-231 cancer cells whereas MCF-10A cell proliferation was not promoted. MCF-10A cell proliferation was promoted on the matrices derived from MCF-10A cells. Chemoresistance of MDA-MB-231 cells against 5-fluorouracil increased on only matrices derived from MDA-MB-231 cells. Our results showed that the cells showed different behaviors on staged tumorigenesis-mimicking matrices according to the malignancy of cell sources for ECM preparation. Therefore, staged tumorigenesis-mimicking matrices might be a useful in vitro ECM models to investigate the roles of ECM in tumor progression.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan; International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | | |
Collapse
|