1
|
Das A, Giri S, Dey P. Cell-cell junctional proteins in cancer. Adv Clin Chem 2024; 125:93-142. [PMID: 39988409 DOI: 10.1016/bs.acc.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
A hallmark change during carcinogenesis is disruption or dysregulation of cell-cell junctions. It enables a transformed cell to adopt mesenchymal phenotype and acquire higher potential to migrate and invade. This ultimately leads to cancer metastasis. During this process, junctional proteins undergo remarkable changes in terms of their expressional pattern, localization, and activity. De-localized junctional proteins may adopt atypical roles which might act to either suppress tumorigenesis or facilitate cancer development, depending on several factors. In this chapter, the authors attempt to know the expression pattern of junctional proteins in different types of cancer, understand its significance, and gather knowledge about the mechanisms by which they regulate tumorigenesis and cancer development.
Collapse
Affiliation(s)
- Aparajita Das
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Sarbani Giri
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India.
| | - Pubali Dey
- Molecular and Cell Biology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| |
Collapse
|
2
|
Li WF, Atalla E, Dong J, Konopleva M. BCL2i-Based Therapies and Emerging Resistance in Chronic Lymphocytic Leukemia. Cells 2024; 13:1922. [PMID: 39594670 PMCID: PMC11592612 DOI: 10.3390/cells13221922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Overexpression of the anti-apoptotic protein BCL-2 is a key factor in the pathogenesis of chronic lymphocytic leukemia (CLL) and is associated with poor clinical outcomes. Therapeutic activation of apoptosis in cancer cells using the BCL-2 inhibitor (BCL2i) venetoclax has shown remarkable efficacy in clinical trials, both as monotherapy and combination regimens. However, patients with CLL experience a highly variable clinical course, facing significant challenges in advanced stages due to disease relapse and the emergence of resistant clones. Resistance mechanisms include acquired BCL-2 mutations, alteration of pro-apoptotic and anti-apoptotic proteins, metabolic reprogramming, epigenetic changes, and aberrant signaling pathways. To address this complex disease and improve progression-free survival, strategies targeting multiple signaling pathways and mechanisms have been explored. Randomized clinical trials of venetoclax in combination with Bruton tyrosine kinase (BTK) inhibitors or CD20 monoclonal antibodies have significantly outperformed traditional chemoimmunotherapy in both treatment-naïve and relapsed patients, achieving undetectable minimal residual disease (uMRD) and durable remissions. This review explores the intricate balance between BCL-2 family proteins and their role in the intrinsic apoptosis pathway, discusses venetoclax resistance mechanisms, and highlights the evolving role of venetoclax and other BCL2i-based combination therapies in CLL treatment.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Drug Resistance, Neoplasm/drug effects
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/therapeutic use
- Sulfonamides/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Wing Fai Li
- Department of Internal Medicine, Jacobi Medical Center, Bronx, NY 10461, USA;
| | - Eleftheria Atalla
- Department of Hematology and Oncology, The University of Texas at San Antonio, San Antonio, TX 78249, USA;
| | - Jiaxin Dong
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Marina Konopleva
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|
3
|
Irfan M, Javed Z, Khan K, Khan N, Docea AO, Calina D, Sharifi-Rad J, Cho WC. Apoptosis evasion via long non-coding RNAs in colorectal cancer. Cancer Cell Int 2022; 22:280. [PMID: 36076273 PMCID: PMC9461221 DOI: 10.1186/s12935-022-02695-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/31/2022] [Indexed: 01/03/2023] Open
Abstract
Long non-coding RNA (LncRNA) is a novel and diverse class of regulatory transcripts that are frequently dysregulated in numerous tumor types. LncRNAs are involved in a complicated molecular network, regulating gene expression, and modulating diverse cellular activities in different cancers including colorectal cancer (CRC). Evidence indicates that lncRNAs can be used as a potential biomarker for the prognosis and diagnosis of CRC as they are aberrantly expressed in CRC cells. The high expression or silencing of lncRNAs is associated with cell proliferation, invasion, metastasis, chemoresistance and apoptosis in CRC. LncRNAs exert both pro-apoptotic and anti-apoptotic functions in CRC. The expression of some oncogene lncRNAs is upregulated which leads to the inhibition of apoptotic pathways, similarly, the tumor suppressor lncRNAs are downregulated in CRC. In this review, we describe the function and mechanisms of lncRNAs to regulate the expression of genes that are involved directly or indirectly in controlling cellular apoptosis in CRC. Furthermore, we also discussed the different apoptotic pathways in normal cells and the mechanisms by which CRC evade apoptosis.
Collapse
Affiliation(s)
- Muhammad Irfan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Zeeshan Javed
- Office for Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Khushbukhat Khan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Naila Khan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| |
Collapse
|
4
|
Cao H, Wang D, Gao R, Li C, Feng Y, Chen L. Therapeutic targets and signaling pathways of active components of QiLing decoction against castration-resistant prostate cancer based on network pharmacology. PeerJ 2022; 10:e13481. [PMID: 35782093 PMCID: PMC9245566 DOI: 10.7717/peerj.13481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 05/02/2022] [Indexed: 01/17/2023] Open
Abstract
QiLing decoction (QLD) is a traditional Chinese medicine compound. This study aims to explore the therapeutic effect of QLD in castration-resistant prostate cancer (CRPC) and its potential bio-targets. A total of 51 active components and QLD 149 targets were identified using bioinformatics analysis. Additionally, five optimal hub target genes were screened including tumor protein P53 (TP53), interleukin-6 (IL-6), vascular endothelial growth factor-A (VEGF-A), caspase-3 (CASP-3), and estrogen receptor-1 (ESR-1). The interrelated network between active components of QLD and their potential targets was constructed. The molecular function, biological processes, and signaling pathways of QLD-against CRPC were identified. Moreover, QLD was found to efficiently exert a repressive effect on CRPC tumor growth mainly by suppressing the activation of HIF-α/VEGFA and TNF-α/IL6 signaling pathways, and increasing the P53 expression level. These results successfully indicated the potential anti-CRPC mechanism of the active components of QLD.
Collapse
Affiliation(s)
- Hongwen Cao
- Urology, LONGHUA Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Wang
- Urology, LONGHUA Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Renjie Gao
- Urology, LONGHUA Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenggong Li
- Andrology of Urology, Linshu Hospital of Traditional Chinese Medicine, Linyi, China
| | - Yigeng Feng
- Urology, LONGHUA Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Chen
- Urology, LONGHUA Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Adhikari S, Bhattacharya A, Adhikary S, Singh V, Gadad S, Roy S, Das C. The paradigm of drug resistance in cancer: an epigenetic perspective. Biosci Rep 2022; 42:BSR20211812. [PMID: 35438143 PMCID: PMC9069444 DOI: 10.1042/bsr20211812] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Innate and acquired resistance towards the conventional therapeutic regimen imposes a significant challenge for the successful management of cancer for decades. In patients with advanced carcinomas, acquisition of drug resistance often leads to tumor recurrence and poor prognosis after the first therapeutic cycle. In this context, cancer stem cells (CSCs) are considered as the prime drivers of therapy resistance in cancer due to their 'non-targetable' nature. Drug resistance in cancer is immensely influenced by different properties of CSCs such as epithelial-to-mesenchymal transition (EMT), a profound expression of drug efflux pump genes, detoxification genes, quiescence, and evasion of apoptosis, has been highlighted in this review article. The crucial epigenetic alterations that are intricately associated with regulating different mechanisms of drug resistance, have been discussed thoroughly. Additionally, special attention is drawn towards the epigenetic mechanisms behind the interaction between the cancer cells and their microenvironment which assists in tumor progression and therapy resistance. Finally, we have provided a cumulative overview of the alternative treatment strategies and epigenome-modifying therapies that show the potential of sensitizing the resistant cells towards the conventional treatment strategies. Thus, this review summarizes the epigenetic and molecular background behind therapy resistance, the prime hindrance of present day anti-cancer therapies, and provides an account of the novel complementary epi-drug-based therapeutic strategies to combat drug resistance.
Collapse
Affiliation(s)
- Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Shrikanth S. Gadad
- Department of Molecular and Translational Medicine, Center of Emphasis in Cancer, Texas Tech University Health Sciences Center El Paso, El Paso, TX, U.S.A
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX 78229, U.S.A
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| |
Collapse
|
6
|
Bildik G, Liang X, Sutton MN, Bast RC, Lu Z. DIRAS3: An Imprinted Tumor Suppressor Gene that Regulates RAS and PI3K-driven Cancer Growth, Motility, Autophagy, and Tumor Dormancy. Mol Cancer Ther 2022; 21:25-37. [PMID: 34667114 DOI: 10.1158/1535-7163.mct-21-0331] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/20/2021] [Accepted: 10/11/2021] [Indexed: 01/14/2023]
Abstract
DIRAS3 is an imprinted tumor suppressor gene that encodes a 26 kDa GTPase with 60% amino acid homology to RAS, but with a distinctive 34 amino acid N-terminal extension required to block RAS function. DIRAS3 is maternally imprinted and expressed only from the paternal allele in normal cells. Loss of expression can occur in a single "hit" through multiple mechanisms. Downregulation of DIRAS3 occurs in cancers of the ovary, breast, lung, prostate, colon, brain, and thyroid. Reexpression of DIRAS3 inhibits signaling through PI3 kinase/AKT, JAK/STAT, and RAS/MAPK, blocking malignant transformation, inhibiting cancer cell growth and motility, and preventing angiogenesis. DIRAS3 is a unique endogenous RAS inhibitor that binds directly to RAS, disrupting RAS dimers and clusters, and preventing RAS-induced transformation. DIRAS3 is essential for autophagy and triggers this process through multiple mechanisms. Reexpression of DIRAS3 induces dormancy in a nu/nu mouse xenograft model of ovarian cancer, inhibiting cancer cell growth and angiogenesis. DIRAS3-mediated induction of autophagy facilitates the survival of dormant cancer cells in a nutrient-poor environment. DIRAS3 expression in dormant, drug-resistant autophagic cancer cells can serve as a biomarker and as a target for novel therapy to eliminate the residual disease that remains after conventional therapy.
Collapse
Affiliation(s)
- Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaowen Liang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Margie N Sutton
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
7
|
Ozyerli-Goknar E, Bagci-Onder T. Epigenetic Deregulation of Apoptosis in Cancers. Cancers (Basel) 2021; 13:3210. [PMID: 34199020 PMCID: PMC8267644 DOI: 10.3390/cancers13133210] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer cells possess the ability to evade apoptosis. Genetic alterations through mutations in key genes of the apoptotic signaling pathway represent a major adaptive mechanism of apoptosis evasion. In parallel, epigenetic changes via aberrant modifications of DNA and histones to regulate the expression of pro- and antiapoptotic signal mediators represent a major complementary mechanism in apoptosis regulation and therapy response. Most epigenetic changes are governed by the activity of chromatin modifying enzymes that add, remove, or recognize different marks on histones and DNA. Here, we discuss how apoptosis signaling components are deregulated at epigenetic levels, particularly focusing on the roles of chromatin-modifying enzymes in this process. We also review the advances in cancer therapies with epigenetic drugs such as DNMT, HMT, HDAC, and BET inhibitors, as well as their effects on apoptosis modulation in cancer cells. Rewiring the epigenome by drug interventions can provide therapeutic advantage for various cancers by reverting therapy resistance and leading cancer cells to undergo apoptotic cell death.
Collapse
Affiliation(s)
- Ezgi Ozyerli-Goknar
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Istanbul 34450, Turkey;
- Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Istanbul 34450, Turkey;
- Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
8
|
Li J, Xie Y, Wang X, Jiang C, Yuan X, Zhang A, Yang L, Liu C, Zou H, Li F, Hu J. Identification of hub genes associated with esophageal cancer progression using bioinformatics analysis. Oncol Lett 2020; 20:214. [PMID: 32963620 PMCID: PMC7491038 DOI: 10.3892/ol.2020.12077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/23/2020] [Indexed: 01/02/2023] Open
Abstract
The underlying causes of esophageal cancer (EC) are unknown. To explore the molecular mechanisms that lead to EC, gene expression profiles of large cohorts of patients with EC were obtained from The Cancer Genome Atlas and the Gene Expression Omnibus (GEO) databases (GSE5364, GSE20347 and GSE23400). The present study identified 83 upregulated and 22 downregulated genes between EC and normal tissue using R statistical software and the GEO2R web tool. The Database for Annotation, Visualization and Integrated Discovery was used to identify the associated pathways, and for functional annotation of the differentially expressed genes (DEGs). Protein-protein interactions of these DEGs were analyzed based on the Search Tool for the Retrieval of Interacting Genes database, and hub genes were visualized using Cytoscape software. An online Kaplan-Meier plotter survival analysis tool was utilized to evaluate the prognostic value of hub gene expression in patients with EC. Further analysis of an additional dataset from GEO (GSE21293) revealed that these genes were associated with infiltration and metastasis in EC. In addition, the Gene Expression Profiling Interactive Analysis tool was used to evaluate expression levels of hub genes in patients with EC for different pathological stages. The Ualcan analysis tool was used to evaluate the expression levels of hub genes for different histological types. Overall, ubiquitin conjugating enzyme E2 C, cyclin dependent kinase inhibitor 3, CDC28 protein kinase regulatory subunit 2, kinesin family member 20A (KIF20A) and RAD51 associated protein 1 (RAD51AP1) were upregulated in EC tissues compared with normal tissues, and upregulation of these genes was a poor prognostic factor for patients with EC, indicating that these genes may mediate EC cell infiltration and metastasis. Among the hub genes, KIF-20A had potential value for predicting the pathological stage of EC. KIF20A and RAD51AP1 were more informative biomarkers of esophageal squamous cell carcinoma. Further studies are required to explore the value of these genes in the treatment of EC.
Collapse
Affiliation(s)
- Jiangfen Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Yufang Xie
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Xueli Wang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Chenhao Jiang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Xin Yuan
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Anzhi Zhang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Lan Yang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Chunxia Liu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Hong Zou
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Feng Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100000, P.R. China
| | - Jianming Hu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| |
Collapse
|
9
|
Bridges AE, Ramachandran S, Pathania R, Parwal U, Lester A, Rajpurohit P, Morera DS, Patel N, Singh N, Korkaya H, Manicassamy S, Prasad PD, Lokeshwar VB, Lokeshwar BL, Ganapathy V, Thangaraju M. RAD51AP1 Deficiency Reduces Tumor Growth by Targeting Stem Cell Self-Renewal. Cancer Res 2020; 80:3855-3866. [PMID: 32665355 DOI: 10.1158/0008-5472.can-19-3713] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/31/2020] [Accepted: 07/09/2020] [Indexed: 11/16/2022]
Abstract
RAD51-associated protein 1 (RAD51AP1) plays an integral role in homologous recombination by activating RAD51 recombinase. Homologous recombination is essential for preserving genome integrity and RAD51AP1 is critical for D-loop formation, a key step in homologous recombination. Although RAD51AP1 is involved in maintaining genomic stability, recent studies have shown that RAD51AP1 expression is significantly upregulated in human cancers. However, the functional role of RAD51AP1 in tumor growth and the underlying molecular mechanism(s) by which RAD51AP1 regulates tumorigenesis have not been fully understood. Here, we use Rad51ap1-knockout mice in genetically engineered mouse models of breast cancer to unravel the role of RAD51AP1 in tumor growth and metastasis. RAD51AP1 gene transcript was increased in both luminal estrogen receptor-positive breast cancer and basal triple-negative breast cancer, which is associated with poor prognosis. Conversely, knockdown of RAD51AP1 (RADP51AP1 KD) in breast cancer cell lines reduced tumor growth. Rad51ap1-deficient mice were protected from oncogene-driven spontaneous mouse mammary tumor growth and associated lung metastasis. In vivo, limiting dilution studies provided evidence that Rad51ap1 plays a critical role in breast cancer stem cell (BCSC) self-renewal. RAD51AP1 KD improved chemotherapy and radiotherapy response by inhibiting BCSC self-renewal and associated pluripotency. Overall, our study provides genetic and biochemical evidences that RAD51AP1 is critical for tumor growth and metastasis by increasing BCSC self-renewal and may serve as a novel target for chemotherapy- and radiotherapy-resistant breast cancer. SIGNIFICANCE: This study provides in vivo evidence that RAD51AP1 plays a critical role in breast cancer growth and metastasis by regulating breast cancer stem cell self-renewal.
Collapse
Affiliation(s)
- Allison E Bridges
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Sabarish Ramachandran
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Rajneesh Pathania
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Utkarsh Parwal
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Adrienne Lester
- Depatment of Undergraduate Health Professions, College of Allied Health Sciences, Augusta University, Augusta, Georgia
| | - Pragya Rajpurohit
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Daley S Morera
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Nikhil Patel
- Department of Pathology, Augusta University, Augusta, Georgia
| | - Nagendra Singh
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Hasan Korkaya
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Santhakumar Manicassamy
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Puttur D Prasad
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Vinata B Lokeshwar
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Bal L Lokeshwar
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia. .,Georgia Cancer Center Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
10
|
Critselis E, Rava M, Marquez M, Lygirou V, Chatzicharalambous D, Liapi P, Lichtinghagen R, Brand K, Cecchini L, Vlahou A, Malats N, Zoidakis J. Diagnostic and Prognostic Performance of Secreted Protein Acidic and Rich in Cysteine (SPARC) Assay for Detecting Primary and Recurrent Urinary Bladder Cancer. Proteomics Clin Appl 2019; 13:e1800148. [DOI: 10.1002/prca.201800148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/03/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Elena Critselis
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Marta Rava
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Mirari Marquez
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Vasiliki Lygirou
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Despoina Chatzicharalambous
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Panagiota Liapi
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Ralph Lichtinghagen
- Institute of Clinical ChemistryHannover Medical School Hannover 30625 Germany
| | - Korbinian Brand
- Institute of Clinical ChemistryHannover Medical School Hannover 30625 Germany
| | | | - Antonia Vlahou
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Nuria Malats
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Jerome Zoidakis
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| |
Collapse
|
11
|
Niture S, Dong X, Arthur E, Chimeh U, Niture SS, Zheng W, Kumar D. Oncogenic Role of Tumor Necrosis Factor α-Induced Protein 8 (TNFAIP8). Cells 2018; 8:cells8010009. [PMID: 30586922 PMCID: PMC6356598 DOI: 10.3390/cells8010009] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor necrosis factor (TNF)-α-induced protein 8 (TNFAIP8) is a founding member of the TIPE family, which also includes TNFAIP8-like 1 (TIPE1), TNFAIP8-like 2 (TIPE2), and TNFAIP8-like 3 (TIPE3) proteins. Expression of TNFAIP8 is strongly associated with the development of various cancers including cancer of the prostate, liver, lung, breast, colon, esophagus, ovary, cervix, pancreas, and others. In human cancers, TNFAIP8 promotes cell proliferation, invasion, metastasis, drug resistance, autophagy, and tumorigenesis by inhibition of cell apoptosis. In order to better understand the molecular aspects, biological functions, and potential roles of TNFAIP8 in carcinogenesis, in this review, we focused on the expression, regulation, structural aspects, modifications/interactions, and oncogenic role of TNFAIP8 proteins in human cancers.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Xialan Dong
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | | | - Weifan Zheng
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
12
|
Fang Y, Shen X. Ubiquitin carboxyl-terminal hydrolases: involvement in cancer progression and clinical implications. Cancer Metastasis Rev 2018; 36:669-682. [PMID: 29080080 DOI: 10.1007/s10555-017-9702-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein ubiquitination and deubiquitination participate in a number of biological processes, including cell growth, differentiation, transcriptional regulation, and oncogenesis. Ubiquitin C-terminal hydrolases (UCHs), a subfamily of deubiquitinating enzymes (DUBs), includes four members: UCH-L1/PGP9.5 (protein gene product 9.5), UCH-L3, UCHL5/UCH37, and BRCA1-associated protein-1 (BAP1). Recently, more attention has been paid to the relationship between the UCH family and malignancies, which play different roles in the progression of different tumors. It remains controversial whether UCHL1 is a tumor promoter or suppressor. UCHL3 and UCH37 are considered to be tumor promoters, while BAP1 is considered to be a tumor suppressor. Studies have showed that UCH enzymes influence several signaling pathways that play crucial roles in oncogenesis, tumor invasion, and migration. In addition, UCH families are associated with tumor cell sensitivity to therapeutic modalities. Here, we reviewed the roles of UCH enzymes in the development of tumors, highlighting the potential consideration of UCH enzymes as new interesting targets for the development of anticancer drugs.
Collapse
Affiliation(s)
- Ying Fang
- The Department of Gastroenterology of Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, People's Republic of China
| | - Xizhong Shen
- The Department of Gastroenterology of Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, People's Republic of China. .,Key Laboratory of Medical Molecule Virology, Ministry of Education and Health, Shanghai Institute of Liver Diseases Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
13
|
Zhang L, Liu R, Luan YY, Yao YM. Tumor Necrosis Factor-α Induced Protein 8: Pathophysiology, Clinical Significance, and Regulatory Mechanism. Int J Biol Sci 2018; 14:398-405. [PMID: 29725261 PMCID: PMC5930472 DOI: 10.7150/ijbs.23268] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-α-induced protein-8 (TNFAIP8) is the earliest discovered component of TNFAIP8 family [tumor necrosis factor-α-induced protein-8 like (TIPE) family]. TNFAIP8 contains a putative death effector domain (DED) homologous to DED II in FLIP (Fas-associated death domain-like interleukin-1β-converting enzyme-inhibitory protein), which may affect cell survival/death process. Recently, it has been demonstrated that TNFAIP8 could inhibit apoptosis and autophagy in various types of cells. Moreover, TNFAIP8 level fluctuated evidently in patients with inflammatory, malignant, and autoimmune diseases, indicating that it might be an anti-apoptotic and oncogenetic protein. Herein we will review the discovery, gene/protein structure, pathophysiological functions, and clinical significance of TNFAIP8 together with its potential regulatory mechanism.
Collapse
Affiliation(s)
- Lei Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.,Emergency Department, The General Hospital of the Chinese PLA Rocket Force, Beijing 100088, People's Republic of China
| | - Ran Liu
- Department of Endocrinology, 307th Hospital of the Chinese PLA, Beijing 100071, People's Republic of China
| | - Ying-Yi Luan
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| |
Collapse
|
14
|
Said N. Roles of SPARC in urothelial carcinogenesis, progression and metastasis. Oncotarget 2018; 7:67574-67585. [PMID: 27564266 PMCID: PMC5341897 DOI: 10.18632/oncotarget.11590] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022] Open
Abstract
Secreted Protein Acidic and Rich in Cysteine (SPARC) is a matricellular glycoprotein that is implicated in myriad physiological and pathological conditions characterized by extensive remodeling and plasticity. The functions and disease association of SPARC in cancer is being increasingly appreciated as it plays multi-faceted contextual roles depending on the cancer type, cell of origin and the unique cancer milieu at both primary and metastatic sites. Herein we will review our current knowledge of the role of SPARC in the multistep cascades of urinary bladder carcinogenesis, progression and metastasis from preclinical models and clinical data and shine the light on its prognostic and therapeutic potentials.
Collapse
Affiliation(s)
- Neveen Said
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston Salem, NC, USA
| |
Collapse
|
15
|
Pires E, Sung P, Wiese C. Role of RAD51AP1 in homologous recombination DNA repair and carcinogenesis. DNA Repair (Amst) 2017; 59:76-81. [PMID: 28963981 PMCID: PMC5643253 DOI: 10.1016/j.dnarep.2017.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/01/2017] [Accepted: 09/21/2017] [Indexed: 12/11/2022]
Abstract
Homologous recombination (HR) serves to repair DNA double-strand breaks and damaged replication forks and is essential for maintaining genome stability and tumor suppression. HR capacity also determines the efficacy of anticancer therapy. Hence, there is an urgent need to better understand all HR proteins and sub-pathways. An emerging protein that is critical for RAD51-mediated HR is RAD51-associated protein 1 (RAD51AP1). Although much has been learned about its biochemical attributes, the precise molecular role of RAD51AP1 in the HR reaction is not yet fully understood. The available literature also suggests that RAD51AP1 expression may be relevant for cancer development and progression. Here, we review the efforts that led to the discovery of RAD51AP1 and elaborate on our current understanding of its biochemical profile and biological function. We also discuss how RAD51AP1 may help to promote cancer development and why it could potentially represent a promising new target for therapeutic intervention.
Collapse
Affiliation(s)
- Elena Pires
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Patrick Sung
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Claudia Wiese
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
16
|
Martín-Subero JI, Esteller M. Epigenetic Mechanisms in Cancer Development. THE MOLECULAR BASIS OF HUMAN CANCER 2017:263-275. [DOI: 10.1007/978-1-59745-458-2_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Gonzalez-Mariscal L, Miranda J, Ortega-Olvera JM, Gallego-Gutierrez H, Raya-Sandino A, Vargas-Sierra O. Zonula Occludens Proteins in Cancer. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0109-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Trevino V, Cassese A, Nagy Z, Zhuang X, Herbert J, Antzack P, Clarke K, Davies N, Rahman A, Campbell MJ, Guindani M, Bicknell R, Vannucci M, Falciani F. A Network Biology Approach Identifies Molecular Cross-Talk between Normal Prostate Epithelial and Prostate Carcinoma Cells. PLoS Comput Biol 2016; 12:e1004884. [PMID: 27124473 PMCID: PMC4849722 DOI: 10.1371/journal.pcbi.1004884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/24/2016] [Indexed: 11/19/2022] Open
Abstract
The advent of functional genomics has enabled the genome-wide characterization of the molecular state of cells and tissues, virtually at every level of biological organization. The difficulty in organizing and mining this unprecedented amount of information has stimulated the development of computational methods designed to infer the underlying structure of regulatory networks from observational data. These important developments had a profound impact in biological sciences since they triggered the development of a novel data-driven investigative approach. In cancer research, this strategy has been particularly successful. It has contributed to the identification of novel biomarkers, to a better characterization of disease heterogeneity and to a more in depth understanding of cancer pathophysiology. However, so far these approaches have not explicitly addressed the challenge of identifying networks representing the interaction of different cell types in a complex tissue. Since these interactions represent an essential part of the biology of both diseased and healthy tissues, it is of paramount importance that this challenge is addressed. Here we report the definition of a network reverse engineering strategy designed to infer directional signals linking adjacent cell types within a complex tissue. The application of this inference strategy to prostate cancer genome-wide expression profiling data validated the approach and revealed that normal epithelial cells exert an anti-tumour activity on prostate carcinoma cells. Moreover, by using a Bayesian hierarchical model integrating genetics and gene expression data and combining this with survival analysis, we show that the expression of putative cell communication genes related to focal adhesion and secretion is affected by epistatic gene copy number variation and it is predictive of patient survival. Ultimately, this study represents a generalizable approach to the challenge of deciphering cell communication networks in a wide spectrum of biological systems. In the current era of cancer research, stimulated by the release of the entire human genome, it has become increasingly clear that to understand cancer we need to understand how the many thousands of genes and proteins involved interact. Modern techniques have enabled the collection of unprecedented amounts of high quality data describing the state of these molecules during cancer development. In cancer research particularly, this strategy has been particularly successful, leading to the discovery of new drugs able to target key factors promoting cancer growth. However, a large body of research suggests that in complex organs, the interaction between cancer and its surrounding environment is an essential part of the biology of both diseased and healthy tissues, therefore it is of paramount importance that this process is further investigated. Here we report a strategy designed to reveal communication signals between cancer cells and adjacent cell types. We apply the strategy to prostate cancer and find that normal cells surrounding the tumour do exert an anti-tumour activity on prostate cancer cells. By using a statistical model which integrates multiple levels of genetic data, we show that cell-to-cell communication genes are controlled by DNA alterations and have potential prognostic value.
Collapse
Affiliation(s)
- Victor Trevino
- Catedra de Bioinformatica, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
| | - Alberto Cassese
- Department of Methodology and Statistics, Maastricht University, Maastricht, Netherlands
| | - Zsuzsanna Nagy
- School of Experimental and Clinical Medicine, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Xiaodong Zhuang
- School of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - John Herbert
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Philipp Antzack
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Kim Clarke
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nicholas Davies
- School of Cancer Sciences, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Ayesha Rahman
- School of Pharmacy, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom
| | - Moray J. Campbell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Michele Guindani
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Roy Bicknell
- School of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Marina Vannucci
- Department of Statistics, Rice University, Houston, Texas, United States of America
| | - Francesco Falciani
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
Pathania R, Ramachandran S, Mariappan G, Thakur P, Shi H, Choi JH, Manicassamy S, Kolhe R, Prasad PD, Sharma S, Lokeshwar BL, Ganapathy V, Thangaraju M. Combined Inhibition of DNMT and HDAC Blocks the Tumorigenicity of Cancer Stem-like Cells and Attenuates Mammary Tumor Growth. Cancer Res 2016; 76:3224-35. [PMID: 27197203 DOI: 10.1158/0008-5472.can-15-2249] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/17/2016] [Indexed: 02/07/2023]
Abstract
Recently, impressive technical advancements have been made in the isolation and validation of mammary stem cells and cancer stem cells (CSC), but the signaling pathways that regulate stem cell self-renewal are largely unknown. Furthermore, CSCs are believed to contribute to chemo- and radioresistance. In this study, we used the MMTV-Neu-Tg mouse mammary tumor model to identify potential new strategies for eliminating CSCs. We found that both luminal progenitor and basal stem cells are susceptible to genetic and epigenetic modifications, which facilitate oncogenic transformation and tumorigenic potential. A combination of the DNMT inhibitor 5-azacytidine and the HDAC inhibitor butyrate markedly reduced CSC abundance and increased the overall survival in this mouse model. RNA-seq analysis of CSCs treated with 5-azacytidine plus butyrate provided evidence that inhibition of chromatin modifiers blocks growth-promoting signaling molecules such as RAD51AP1 and SPC25, which play key roles in DNA damage repair and kinetochore assembly. Moreover, RAD51AP1 and SPC25 were significantly overexpressed in human breast tumor tissues and were associated with reduced overall patient survival. In conclusion, our studies suggest that breast CSCs are intrinsically sensitive to genetic and epigenetic modifications and can therefore be significantly affected by epigenetic-based therapies, warranting further investigation of combined DNMT and HDAC inhibition in refractory or drug-resistant breast cancer. Cancer Res; 76(11); 3224-35. ©2016 AACR.
Collapse
Affiliation(s)
- Rajneesh Pathania
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Sabarish Ramachandran
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Gurusamy Mariappan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Priyanka Thakur
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Huidong Shi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia. CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jeong-Hyeon Choi
- CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. Department of Biostatistics, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Santhakumar Manicassamy
- CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. Immunotherapy Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ravindra Kolhe
- CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Puttur D Prasad
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia. CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Suash Sharma
- CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Bal L Lokeshwar
- CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. Charlie Norwood VA Medical Center and Department of Medicine and Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia. CRU Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
20
|
Liu W, Liu Y, Liu H, Zhang W, Fu Q, Xu J, Gu J. Tumor Suppressive Function of p21-activated Kinase 6 in Hepatocellular Carcinoma. J Biol Chem 2015; 290:28489-28501. [PMID: 26442588 DOI: 10.1074/jbc.m115.658237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Indexed: 01/16/2023] Open
Abstract
Our previous studies identified the oncogenic role of p21-activated kinase 1 (PAK1) in hepatocellular carcinoma (HCC) and renal cell carcinoma (RCC). Contrarily, PAK6 was found to predict a favorable prognosis in RCC patients. Nevertheless, the ambiguous tumor suppressive function of PAK6 in hepatocarcinogenesis remains obscure. Herein, decreased PAK6 expression was found to be associated with tumor node metastasis stage progression and unfavorable overall survival in HCC patients. Additionally, overexpression and silence of PAK6 experiments showed that PAK6 inhibited xenografted tumor growth in vivo, and restricted cell proliferation, colony formation, migration, and invasion and promoted cell apoptosis and anoikis in vitro. Moreover, overexpression of kinase dead and nuclear localization signal deletion mutants of PAK6 experiments indicated the tumor suppressive function of PAK6 was partially dependent on its kinase activity and nuclear translocation. Furthermore, gain or loss of function in polycomb repressive complex 2 (PRC2) components, including EZH2, SUZ12, and EED, elucidated epigenetic control of H3K27me3-arbitrated PAK6 down-regulation in hepatoma cells. More importantly, negative correlation between PAK6 and EZH2 expression was observed in hepatoma tissues from HCC patients. These data identified the tumor suppressive role and potential underlying mechanism of PAK6 in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Weisi Liu
- Departments of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032
| | - Yidong Liu
- Departments of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
| | - Weijuan Zhang
- Departments of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032
| | - Qiang Fu
- Departments of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032
| | - Jiejie Xu
- Departments of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032.
| | - Jianxin Gu
- Departments of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032
| |
Collapse
|
21
|
Lipka DB, Wang Q, Cabezas-Wallscheid N, Klimmeck D, Weichenhan D, Herrmann C, Lier A, Brocks D, von Paleske L, Renders S, Wünsche P, Zeisberger P, Gu L, Haas S, Essers MA, Brors B, Eils R, Trumpp A, Milsom MD, Plass C. Identification of DNA methylation changes at cis-regulatory elements during early steps of HSC differentiation using tagmentation-based whole genome bisulfite sequencing. Cell Cycle 2015; 13:3476-87. [PMID: 25483069 DOI: 10.4161/15384101.2014.973334] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Epigenetic alterations during cellular differentiation are a key molecular mechanism which both instructs and reinforces the process of lineage commitment. Within the haematopoietic system, progressive changes in the DNA methylome of haematopoietic stem cells (HSCs) are essential for the effective production of mature blood cells. Inhibition or loss of function of the cellular DNA methylation machinery has been shown to lead to a severe perturbation in blood production and is also an important driver of malignant transformation. HSCs constitute a very rare cell population in the bone marrow, capable of life-long self-renewal and multi-lineage differentiation. The low abundance of HSCs has been a major technological barrier to the global analysis of the CpG methylation status within both HSCs and their immediate progeny, the multipotent progenitors (MPPs). Within this Extra View article, we review the current understanding of how the DNA methylome regulates normal and malignant hematopoiesis. We also discuss the current methodologies that are available for interrogating the DNA methylation status of HSCs and MPPs and describe a new data set that was generated using tagmentation-based whole genome bisulfite sequencing (TWGBS) in order to comprehensively map methylated cytosines using the limited amount of genomic DNA that can be harvested from rare cell populations. Extended analysis of this data set clearly demonstrates the added value of genome-wide sequencing of methylated cytosines and identifies novel important cis-acting regulatory regions that are dynamically remodeled during the first steps of haematopoietic differentiation.
Collapse
Affiliation(s)
- Daniel B Lipka
- a Division of Epigenomics and Cancer Risk Factors , German Cancer Research Center (DKFZ) ; Heidelberg , Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rotondo JC, Bosi S, Bassi C, Ferracin M, Lanza G, Gafà R, Magri E, Selvatici R, Torresani S, Marci R, Garutti P, Negrini M, Tognon M, Martini F. Gene expression changes in progression of cervical neoplasia revealed by microarray analysis of cervical neoplastic keratinocytes. J Cell Physiol 2015; 230:806-812. [PMID: 25205602 DOI: 10.1002/jcp.24808] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/05/2014] [Indexed: 12/28/2022]
Abstract
To evaluate the gene expression changes involved in neoplastic progression of cervical intraepithelial neoplasia. Using microarray analysis, large-scale gene expression profile was carried out on HPV16-CIN2, HPV16-CIN3, and normal cervical keratinocytes derived from two HPV16-CIN2, two HPV-CIN3 lesions, and two corresponding normal cervical tissues, respectively. Differentially expressed genes were analyzed in normal cervical keratinocytes compared with HPV16-CIN2 keratinocytes and in HPV16-CIN2 keratinocytes compared with HPV16-CIN3 keratinocytes; 37 candidate genes with continuously increasing or decreasing expression during CIN progression were identified. One of these genes, phosphoglycerate dehydrogenase, was chosen for further characterization. Quantitative reverse transcription-polymerase chain reaction and immunohistochemical analysis confirmed that expression of phosphoglycerate dehydrogenase consistently increases during progression of CIN toward cancer. Gene expression changes occurring during CIN progression were investigated using microarray analysis, for the first time, in CIN2 and CIN3 keratinocytes naturally infected with HPV16. Phosphoglycerate dehydrogenase is likely to be associated with tumorigenesis and may be a potential prognostic marker for CIN progression.
Collapse
Affiliation(s)
- John Charles Rotondo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, via Fossato di Mortara 64/B, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gu L, Frommel SC, Oakes CC, Simon R, Grupp K, Gerig CY, Bär D, Robinson MD, Baer C, Weiss M, Gu Z, Schapira M, Kuner R, Sültmann H, Provenzano M, Yaspo ML, Brors B, Korbel J, Schlomm T, Sauter G, Eils R, Plass C, Santoro R. BAZ2A (TIP5) is involved in epigenetic alterations in prostate cancer and its overexpression predicts disease recurrence. Nat Genet 2014; 47:22-30. [PMID: 25485837 DOI: 10.1038/ng.3165] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/17/2014] [Indexed: 12/14/2022]
Abstract
Prostate cancer is driven by a combination of genetic and/or epigenetic alterations. Epigenetic alterations are frequently observed in all human cancers, yet how aberrant epigenetic signatures are established is poorly understood. Here we show that the gene encoding BAZ2A (TIP5), a factor previously implicated in epigenetic rRNA gene silencing, is overexpressed in prostate cancer and is paradoxically involved in maintaining prostate cancer cell growth, a feature specific to cancer cells. BAZ2A regulates numerous protein-coding genes and directly interacts with EZH2 to maintain epigenetic silencing at genes repressed in metastasis. BAZ2A overexpression is tightly associated with a molecular subtype displaying a CpG island methylator phenotype (CIMP). Finally, high BAZ2A levels serve as an independent predictor of biochemical recurrence in a cohort of 7,682 individuals with prostate cancer. This work identifies a new aberrant role for the epigenetic regulator BAZ2A, which can also serve as a useful marker for metastatic potential in prostate cancer.
Collapse
Affiliation(s)
- Lei Gu
- 1] Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany. [2] Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sandra C Frommel
- 1] Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland. [2] Molecular Life Science Program, Life Science Zurich Graduate School, University of Zurich, Zurich, Switzerland
| | - Christopher C Oakes
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Grupp
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cristina Y Gerig
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| | - Dominik Bär
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| | - Mark D Robinson
- 1] Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland. [2] Swiss Institute of Bioinformatics (SIB), University of Zurich, Zurich, Switzerland
| | - Constance Baer
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Weiss
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zuguang Gu
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthieu Schapira
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Ruprecht Kuner
- Unit of Cancer Genome Research, German Cancer Research Center (DKFZ) and National Center of Tumour Diseases, Heidelberg, Germany
| | - Holger Sültmann
- Unit of Cancer Genome Research, German Cancer Research Center (DKFZ) and National Center of Tumour Diseases, Heidelberg, Germany
| | - Maurizio Provenzano
- Oncology Research Unit, Division of Urology, University Hospital of Zurich, Zurich, Switzerland
| | | | | | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Korbel
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Thorsten Schlomm
- Martini Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Eils
- 1] Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany. [2] Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, Heidelberg University, Heidelberg, Germany
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raffaella Santoro
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Abstract
PAKs 4, 5 and 6 are members of the group B family of p21-activated kinases. Among this group, PAK4 has been most extensively studied. While it has essential roles in embryonic development, in adults high levels of PAK4 are frequently associated with cancer. PAK4 is overexpressed in a variety of cancers, and the Pak4 gene is amplified in some cancers. PAK4 overexpression is sufficient to cause oncogenic transformation in cells and in mouse models. The tight connection between PAK4 and cancer make it a promising diagnostic tool as well as a potential drug target. The group B PAKs also have important developmental functions. PAK4 is important for many early developmental processes, while PAK5 and PAK6 play roles in learning and memory in mice. This chapter provides an overview of the roles of the group B PAKs in cancer as well as development, and includes a discussion of PAK mediated signaling pathways and cellular functions.
Collapse
Affiliation(s)
- Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research; Department of Chemical Biology; Ernest Mario School of Pharmacy; Rutgers, The State University of New Jersey; Piscataway, NJ USA
| |
Collapse
|
25
|
Lu Z, Yang H, Sutton MN, Yang M, Clarke CH, Liao WSL, Bast RC. ARHI (DIRAS3) induces autophagy in ovarian cancer cells by downregulating the epidermal growth factor receptor, inhibiting PI3K and Ras/MAP signaling and activating the FOXo3a-mediated induction of Rab7. Cell Death Differ 2014; 21:1275-89. [PMID: 24769729 DOI: 10.1038/cdd.2014.48] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 01/31/2023] Open
Abstract
The process of autophagy has been described in detail at the molecular level in normal cells, but less is known of its regulation in cancer cells. Aplasia Ras homolog member I (ARHI; DIRAS3) is an imprinted tumor suppressor gene that is downregulated in multiple malignancies including ovarian cancer. Re-expression of ARHI slows proliferation, inhibits motility, induces autophagy and produces tumor dormancy. Our previous studies have implicated autophagy in the survival of dormant ovarian cancer cells and have shown that ARHI is required for autophagy induced by starvation or rapamycin treatment. Re-expression of ARHI in ovarian cancer cells blocks signaling through the PI3K and Ras/MAP pathways, which, in turn, downregulates mTOR and initiates autophagy. Here we show that ARHI is required for autophagy-meditated cancer cell arrest and ARHI inhibits signaling through PI3K/AKT and Ras/MAP by enhancing internalization and degradation of the epidermal growth factor receptor. ARHI-mediated downregulation of PI3K/AKT and Ras/ERK signaling also decreases phosphorylation of FOXo3a, which sequesters this transcription factor in the nucleus. Nuclear retention of FOXo3a induces ATG4 and MAP-LC3-I, required for maturation of autophagosomes, and also increases the expression of Rab7, required for fusion of autophagosomes with lysosomes. Following the knockdown of FOXo3a or Rab7, autophagolysosome formation was observed but was markedly inhibited, resulting in numerous enlarged autophagosomes. ARHI expression correlates with LC3 expression and FOXo3a nuclear localization in surgical specimens of ovarian cancer. Thus, ARHI contributes to the induction of autophagy through multiple mechanisms in ovarian cancer cells.
Collapse
Affiliation(s)
- Z Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - H Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - M N Sutton
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - M Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - C H Clarke
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - W S-L Liao
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - R C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| |
Collapse
|
26
|
Liu W, Liu H, Liu Y, Xu L, Zhang W, Zhu Y, Xu J, Gu J. Prognostic Significance of p21-activated Kinase 6 Expression in Patients with Clear Cell Renal Cell Carcinoma. Ann Surg Oncol 2014; 21 Suppl 4:S575-83. [DOI: 10.1245/s10434-014-3680-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Indexed: 02/06/2023]
|
27
|
Nagaraju GP, Dontula R, El-Rayes BF, Lakka SS. Molecular mechanisms underlying the divergent roles of SPARC in human carcinogenesis. Carcinogenesis 2014; 35:967-73. [PMID: 24675529 DOI: 10.1093/carcin/bgu072] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Communication between the cell and its surrounding environment, consisting of proteinaceous (non-living material) and extracellular matrix (ECM), is important for biophysiological and chemical signaling. This signaling results in a range of cellular activities, including cell division, adhesion, differentiation, invasion, migration and angiogenesis. The ECM non-structural secretory glycoprotein called secreted protein, acidic and rich in cysteine (SPARC), plays a significant role in altering cancer cell activity and the tumor's microenvironment (TME). However, the role of SPARC in cancer research has been the subject of controversy. This review mainly focuses on recent advances in understanding the contradictory nature of SPARC in relation to ECM assembly, cancer cell proliferation, adhesion, migration, apoptosis and tumor growth.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA and
| | | | | | | |
Collapse
|
28
|
Abstract
The p21 activated kinases (Paks) are well known effector proteins for the Rho GTPases Cdc42 and Rac. The Paks contain 6 members, which fall into 2 families of proteins. The first family consists of Paks 1, 2, and 3, and the second consists of Paks 4, 5, and 6. While some of the Paks are ubiquitously expressed, others have more restrictive tissue specificity. All of them are found in the nervous system. Studies using cell culture, transgenic mice, and knockout mice, have revealed important roles for the Paks in cytoskeletal organization and in many aspects of cell growth and development. This review discusses the basic structures of the Paks, and their roles in cell growth, development, and in cancer.
Collapse
Affiliation(s)
- Chetan K Rane
- Susan Lehman Cullman Laboratory for Cancer Research; Department of Chemical Biology; Ernest Mario School of Pharmacy; Rutgers The State University of New Jersey; Piscataway, NJ USA
| | - Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research; Department of Chemical Biology; Ernest Mario School of Pharmacy; Rutgers The State University of New Jersey; Piscataway, NJ USA
| |
Collapse
|
29
|
Pignatta S, Arienti C, Zoli W, Di Donato M, Castoria G, Gabucci E, Casadio V, Falconi M, De Giorgi U, Silvestrini R, Tesei A. Prolonged exposure to (R)-bicalutamide generates a LNCaP subclone with alteration of mitochondrial genome. Mol Cell Endocrinol 2014; 382:314-324. [PMID: 24397920 DOI: 10.1016/j.mce.2013.10.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/18/2013] [Accepted: 10/18/2013] [Indexed: 01/05/2023]
Abstract
Advanced prostate cancers, initially sensitive to androgen deprivation therapy, frequently progress to the castration-resistant prostate cancer phenotype (CRPC) through mechanisms not yet fully understood. In this study we investigated mitochondrial involvement in the establishment of refractoriness to hormone therapy. Two human prostate cancer cell lines were used, the parental LNCaP and the resistant LNCaP-Rbic, the latter generated after continuous exposure to 20 μM of (R)-bicalutamide, the active enantiomer of Casodex®. We observed a significant decrease in mtDNA content and a lower expression of 8 mitochondria-encoded gene transcripts involved in respiratory chain complexes in both cell lines. We also found that (R)-bicalutamide differentially modulated dynamin-related protein (Drp-1) expression in LNCaP and LNCaP-Rbic cells. These data seem to indicate that the androgen-independent phenotype in our experimental model was due, at least in part, to alterations in mitochondrial dynamics and to a breakdown in the Drp-1-mediated mitochondrial network.
Collapse
Affiliation(s)
- Sara Pignatta
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Arienti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Wainer Zoli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Marzia Di Donato
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Gabriella Castoria
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Elisa Gabucci
- Department of General Surgery and Organ Transplantation, University of Bologna, Italy
| | - Valentina Casadio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Mirella Falconi
- Department of Human Anatomy and Physiopathology of the Locomotor Apparatus, University of Bologna, Italy
| | - Ugo De Giorgi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Rosella Silvestrini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Tesei
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
30
|
Role of p-21-activated kinases in cancer progression. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 309:347-87. [PMID: 24529727 DOI: 10.1016/b978-0-12-800255-1.00007-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The p-21-activated kinases (PAKs) are downstream effectors of Rho GTPases Rac and Cdc42. The PAK family consists of six members which are segregated into two subgroups (Group I and Group II) based on sequence homology. Group I PAKs (PAK1-3) are the most extensively studied but there is increasing interest in the functionality of Group II PAKs (PAK4-6). The PAK family proteins are thought to play an important role in many different cellular processes, some of which have particular significance in the context of cancer progression. This review explores established and more recent data, linking the PAK family kinases to cancer progression including expression profiles, evasion of apoptosis, promotion of cell survival, and regulation of cell invasion. Finally, we discuss attempts to therapeutically target the PAK family and outline the major obstacles that still need to be overcome.
Collapse
|
31
|
Bethge N, Lothe RA, Honne H, Andresen K, Trøen G, Eknæs M, Liestøl K, Holte H, Delabie J, Smeland EB, Lind GE. Colorectal cancer DNA methylation marker panel validated with high performance in Non-Hodgkin lymphoma. Epigenetics 2013; 9:428-36. [PMID: 24362313 PMCID: PMC4053461 DOI: 10.4161/epi.27554] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Genes with altered DNA methylation can be used as biomarkers for cancer detection and assessment of prognosis. Here we analyzed the methylation status of a colorectal cancer biomarker panel (CNRIP1, FBN1, INA, MAL, SNCA, and SPG20) in 97 cancer cell lines, derived from 17 different cancer types. Interestingly, the genes were frequently methylated also in hematological cancer types and were therefore subjected to analyses in primary tumor samples from the major types of non-Hodgkin lymphomas (NHL) and in healthy controls. In total, the genes CNRIP1, FBN1, INA, MAL, SNCA, and SPG20 were methylated in 53%, 23%, 52%, 69%, 97%, and 92% of the tumor samples, respectively, and were unmethylated in all healthy controls. With the exception of a single tumor sample, a correct prediction of lymphoma or normal sample was made in a blinded analysis of the validation series using a combination of SNCA and SPG20. The combined ROC-curve analysis of these genes resulted in an area under the curve of 0.999 (P = 4.2 × 10−18), and a sensitivity and specificity of 98% and 100%, respectively, across the test and validation series. Interestingly, the promoter methylation of CNRIP1 was associated with decreased overall survival in diffuse large B-cell lymphoma (DLBCL) (P = 0.03).
In conclusion, our results demonstrate that SNCA and SPG20 methylation might be suitable for early detection and monitoring of NHL. Furthermore, CNRIP1 could potentially be used as a prognostic factor in DLBCL.
Collapse
Affiliation(s)
- Nicole Bethge
- Department of Immunology; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway; Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway
| | - Ragnhild A Lothe
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Cancer Prevention; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway
| | - Hilde Honne
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Cancer Prevention; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway
| | - Kim Andresen
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Cancer Prevention; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway
| | - Gunhild Trøen
- Department of Pathology; Oslo University Hospital; Oslo, Norway
| | - Mette Eknæs
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Cancer Prevention; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway
| | - Knut Liestøl
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Informatics; University of Oslo; Oslo, Norway
| | - Harald Holte
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Oncology; The Norwegian Radium Hospital; Oslo University Hospital; Oslo, Norway
| | - Jan Delabie
- Department of Pathology; Oslo University Hospital; Oslo, Norway
| | - Erlend B Smeland
- Department of Immunology; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway; Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway
| | - Guro E Lind
- Centre for Cancer Biomedicine; University of Oslo; Oslo, Norway; Department of Cancer Prevention; Institute for Cancer Research; Oslo University Hospital; Oslo, Norway
| |
Collapse
|
32
|
Lyu T, Jia N, Wang J, Yan X, Yu Y, Lu Z, Bast RC, Hua K, Feng W. Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 2013; 8:1330-46. [PMID: 24135786 DOI: 10.4161/epi.26675] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The initiation of angiogenesis can mark the transition from tumor dormancy to active growth and recurrence. Mechanisms that regulate recurrence in human cancers are poorly understood, in part because of the absence of relevant models. The induction of ARHI (DIRAS3) induces dormancy and autophagy in human ovarian cancer xenografts but produces autophagic cell death in culture. The addition of VEGF to cultures maintains the viability of dormant autophagic cancer cells, thereby permitting active growth when ARHI is downregulated, which mimics the "recurrence" of growth in xenografts. Two inducible ovarian cancer cell lines, SKOv3-ARHI and Hey-ARHI, were used. The expression level of angiogenesis factors was evaluated by real-time PCR, immunohistochemistry, immunocytochemistry and western blot; their epigenetic regulation was measured by bisulfite sequencing and chromatin immunoprecipitation. Six of the 15 angiogenesis factors were upregulated in dormant cancer cells (tissue inhibitor of metalloproteinases-3, TIMP3; thrombospondin-1, TSP1; angiopoietin-1; angiopoietin-2; angiopoietin-4; E-cadherin, CDH1). We found that TIMP3 and CDH1 expression was regulated epigenetically and was related inversely to the DNA methylation of their promoters in cell cultures and in xenografts. Increased H3K9 acetylation was associated with higher TIMP3 expression in dormant SKOv3-ARHI cells, while decreased H3K27me3 resulted in the upregulation of TIMP3 in dormant Hey-ARHI cells. Elevated CDH1 expression during dormancy was associated with an increase in both H3K4me3 and H3K9Ac in two cell lines. CpG demethylating agents and/or histone deacetylase inhibitors inhibited the re-growth of dormant cancer cells, which was associated with the re-expression of anti-angiogenic genes. The expression of the anti-angiogenic genes TIMP3 and CDH1 is elevated during dormancy and is reduced during the transition to active growth by changes in DNA methylation and histone modification.
Collapse
Affiliation(s)
- Tianjiao Lyu
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Nan Jia
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Jieyu Wang
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Xiaohui Yan
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Yinhua Yu
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Zhen Lu
- Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Robert C Bast
- Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Keqin Hua
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Weiwei Feng
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| |
Collapse
|
33
|
An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism. PLoS One 2013; 8:e61758. [PMID: 23626723 PMCID: PMC3634018 DOI: 10.1371/journal.pone.0061758] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/11/2013] [Indexed: 11/19/2022] Open
Abstract
Long chain alkylphenols are man-made compounds still present in industrial and agricultural processes. Their main use is domestic and they are widespread in household products, cleansers and cosmetics, leading to a global environmental and human contamination. These molecules are known to exert estrogen-like activities through binding to classical estrogen receptors. In vitro, they can also interact with the G-protein coupled estrogen receptor. Testicular germ cell tumor etiology and progression are proposed to be stimulated by lifelong estrogeno-mimetic exposure. We studied the transduction signaling pathways through which an alkyphenol mixture triggers testicular cancer cell proliferation in vitro and in vivo. Proliferation assays were monitored after exposure to a realistic mixture of 4-tert-octylphenol and 4-nonylphenol of either TCam-2 seminoma derived cells, NT2/D1 embryonal carcinoma cells or testis tumor in xenografted nude mice. Specific pharmacological inhibitors and gene-silencing strategies were used in TCam-2 cells in order to demonstrate that the alkylphenol mix triggers CREB-phosphorylation through a rapid, ERα36-PI3kinase non genomic pathway. Microarray analysis of the mixture target genes revealed that this pathway can modulate the expression of the DNA-methyltransferase-3 (Dnmt3) gene family which is involved in DNA methylation control. Our results highlight a key role for ERα36 in alkylphenol non genomic signaling in testicular germ cell tumors. Hence, ERα36-dependent control of the epigenetic status opens the way for the understanding of the link between endocrine disruptor exposure and the burden of hormone sensitive cancers.
Collapse
|
34
|
Bachet JB, Tabone-Eglinger S, Dessaux S, Besse A, Brahimi-Adouane S, Emile JF, Blay JY, Alberti L. Gene expression patterns of hemizygous and heterozygous KIT mutations suggest distinct oncogenic pathways: a study in NIH3T3 cell lines and GIST samples. PLoS One 2013; 8:e61103. [PMID: 23593401 PMCID: PMC3625162 DOI: 10.1371/journal.pone.0061103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/05/2013] [Indexed: 12/18/2022] Open
Abstract
Objective Most gain of function mutations of tyrosine kinase receptors in human tumours are hemizygous. Gastrointestinal stromal tumours (GIST) with homozygous mutations have a worse prognosis. We aimed to identify genes differentially regulated by hemizygous and heterozygous KIT mutations. Materials and Methods Expression of 94 genes and 384 miRNA was analysed with low density arrays in five NIH3T3 cell lines expressing the full-length human KIT cDNA wild-type (WT), hemizygous KIT mutation with del557-558 (D6) or del564-581 (D54) and heterozygous WT/D6 or WT/D54. Expression of 5 of these genes and 384 miRNA was then analysed in GISTs samples. Results Unsupervised and supervised hierarchical clustering of the mRNA and miRNA profiles showed that heterozygous mutants clustered with KIT WT expressing cells while hemizygous mutants were distinct. Among hemizygous cells, D6 and D54 expressing cells clustered separately. Most deregulated genes have been reported as potentially implicated in cancer and severals, as ANXA8 and FBN1, are highlighted by both, mRNA and miRNA analyses. MiRNA and mRNA analyses in GISTs samples confirmed that their expressions varied according to the mutation of the alleles. Interestingly, RGS16, a membrane protein of the regulator of G protein family, correlate with the subcellular localization of KIT mutants and might be responsible for regulation of the PI3K/AKT signalling pathway. Conclusion Patterns of mRNA and miRNA expression in cells and tumours depend on heterozygous/hemizygous status of KIT mutations, and deletion/presence of TYR568 & TYR570 residues. Thus each mutation of KIT may drive specific oncogenic pathways.
Collapse
Affiliation(s)
- Jean-Baptiste Bachet
- EA4340 'Epidémiologie et Oncogénèse des tumeurs digestives', Faculté de médecine PIFO, UVSQ, Guyancourt, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Trifa F, Karray-Chouayekh S, Jmaa ZB, Jmal E, Khabir A, Sellami-Boudawara T, Frikha M, Daoud J, Mokdad-Gargouri R. Frequent CpG methylation of ubiquitin carboxyl-terminal hydrolase 1 (UCHL1) in sporadic and hereditary Tunisian breast cancer patients: clinical significance. Med Oncol 2013; 30:418. [DOI: 10.1007/s12032-012-0418-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 11/09/2012] [Indexed: 12/29/2022]
|
36
|
Zhang C, Kallakury BV, Ross JS, Mewani RR, Sheehan CE, Sakabe I, Luta G, Kumar D, Yadavalli S, Starr J, Sreenath TL, Srivastava S, Pollard HB, Eidelman O, Srivastava M, Kasid UN. The significance of TNFAIP8 in prostate cancer response to radiation and docetaxel and disease recurrence. Int J Cancer 2013; 133:31-42. [PMID: 23280553 DOI: 10.1002/ijc.27996] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 11/19/2012] [Indexed: 12/12/2022]
Abstract
TNFAIP8 is a NF-κB-inducible, oncogenic molecule. Previous "promoter array" studies have identified differential methylation and regulation of TNFAIP8 in prostate epithelial and cancer cell lines. Here we demonstrate that TNFAIP8 expression is induced by androgen in hormone-responsive LNCaP prostate cancer cells. In athymic mice bearing hormone-refractory PC-3 prostate tumor xenografts, intravenous treatment with a liposomal formulation of TNFAIP8 antisense oligonucleotide (LE-AS5) caused reduced expression of TNFAIP8 in tumor tissues, and a combination of LE-AS5 and radiation or docetaxel treatment resulted in significant inhibition of PC-3 tumor growth as compared to single agents. The immunohistochemical evaluation of TNFAIP8 expression revealed correlation of both cytoplasmic and nuclear TNFAIP8 overexpression with high grade prostatic adenocarcinomas, while nuclear overexpression was found to be an independent predictor of disease recurrence controlling for tumor grade. Increased nuclear TNFAIP8 expression was statistically significantly associated with a 2.44 fold (95 % confidence interval: 1.01-5.91) higher risk of prostate cancer recurrence. Mechanistically, TNFAIP8 seems to function as a scaffold (or adaptor) protein. In the antibody microarray analysis of proteins associated with the TNFAIP8 immune-complex, we have identified Karyopherin alpha2 as a novel binding partner of nuclear TNFAIP8 in PC-3 cells. The Ingenuity Pathway Analysis of the TNFAIP8 interacting proteins suggested that TNFAIP8 influences cancer progression pathways and networks involving integrins and matrix metalloproteinases. Taken together, present studies demonstrate that TNFAIP8 is a novel therapeutic target in prostate cancer, and indicate a potential relationship of the nuclear trafficking of TNFAIP8 with adverse outcomes in a subset of prostate cancer patients.
Collapse
Affiliation(s)
- Chuanbo Zhang
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gonzalez-Mariscal L, Bautista P, Lechuga S, Quiros M. ZO-2, a tight junction scaffold protein involved in the regulation of cell proliferation and apoptosis. Ann N Y Acad Sci 2012; 1257:133-41. [PMID: 22671599 DOI: 10.1111/j.1749-6632.2012.06537.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
ZO-2 is a membrane-associated guanylate kinase homologue (MAGUK) tight protein associated with the cytoplasmic surface of tight junctions. Here, we describe how ZO-2 is a multidomain molecule that binds to a variety of cell signaling proteins, to the actin cytoskeleton, and to gap, tight, and adherens junction proteins. In sparse cultures, ZO-2 is present at the nucleus and associates with molecules active in gene transcription and pre-mRNA processing. ZO-2 inhibits the Wnt signaling pathway, reduces cell proliferation, and promotes apoptosis; its absence, mutation, or overexpression is present in various human diseases, including deafness and cancer.
Collapse
Affiliation(s)
- Lorenza Gonzalez-Mariscal
- Center of Research and Advanced Studies, Cinvestav, Department of Physiology, Biophysics and Neuroscience, Mexico DF, Mexico.
| | | | | | | |
Collapse
|
38
|
Genomic Signatures Associated with the Development, Progression, and Outcome of Prostate Cancer. Mol Diagn Ther 2012; 11:345-54. [DOI: 10.1007/bf03256258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Zhang Y, Yang B, Du Z, Bai T, Gao YT, Wang YJ, Lou C, Wang FM, Bai Y. Aberrant methylation of SPARC in human hepatocellular carcinoma and its clinical implication. World J Gastroenterol 2012; 18:2043-2052. [PMID: 22563191 PMCID: PMC3342602 DOI: 10.3748/wjg.v18.i17.2043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/25/2011] [Accepted: 02/27/2012] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the methylation status of secreted protein acidic and rich in cysteine (SPARC) in human hepatocellular carcinoma (HCC) and evaluate its clinical implication. METHODS The methylation status of SPARC was analyzed in one HCC cell line (SMMC-7721) and 60 pairs of HCC and corresponding nontumorous tissues by methylation-specific polymerase chain reaction and bisulfite sequencing. The expression of SPARC mRNA and protein were examined by reverse transcription polymerase chain reaction and immunohistochemistry, respectively. The correlations between the methylation status and the gene expression, the clinicopathological parameters, as well as the prognosis after surgery were analyzed. RESULTS In the SMMC-7721 cell line, the loss of SPARC expression was correlated with the aberrant methylation and could be reactivated by the demethylating agent 5-aza-2'-deoxycytidine. Methylation frequency of SPARC in HCC was significantly higher than that in the corresponding nontumorous tissues (45/60 vs 7/60, P < 0.001), and it was correlated with the pathological classification (P = 0.019). The downregulation of the SPARC mRNA expression in HCC was correlated with the SPARC methylation (P = 0.040). The patients with methylated SPARC had a poorer overall survival than those without methylated SPARC (28.0 mo vs 41.0 mo, P = 0.043). CONCLUSION Aberrant methylation is an important mechanism for SPARC inactivation in HCC and SPARC methylation may be a promising biomarker for the diagnosis and prognosis of HCC.
Collapse
|
40
|
Adjakly M, Bosviel R, Rabiau N, Boiteux JP, Bignon YJ, Guy L, Bernard-Gallon D. DNA methylation and soy phytoestrogens: quantitative study in DU-145 and PC-3 human prostate cancer cell lines. Epigenomics 2011; 3:795-803. [PMID: 22126297 DOI: 10.2217/epi.11.103] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM DNA hypermethylation is an epigenetic mechanism which induces silencing of tumor-suppressor genes in prostate cancer. Many studies have reported that specific components of food plants like soy phytoestrogens may have protective effects against prostate carcinogenesis or progression. Genistein and daidzein, the major phytoestrogens, have been reported to have the ability to reverse DNA hypermethylation in cancer cell lines. The aim of this study was to investigate the potential demethylating effects of these two soy compounds on BRCA1, GSTP1, EPHB2 and BRCA2 promoter genes. METHODS & MATERIALS Prostate cell lines DU-145 and PC-3 were treated with genistein 40 µM, daidzein 110 µM, budesonide (methylating agent) 2 µM and 5-azacytidine (demethylating agent) 2 µM. In these two human prostate cancer cell lines we performed methylation quantification by using Methyl Profiler DNA methylation analysis. This technique is based on a methylation-specific digestion followed by quantitative PCR. We analyzed the corresponding protein expression by western blotting. RESULTS Soy phytoestrogens induced a demethylation of all promoter regions studied except for BRCA2, which is not methylated in control cell lines. An increase in their protein expression was also demonstrated by western blot analysis and corroborated the potential demethylating effect of soy phytoestrogens. CONCLUSION This study showed that the soy phytoestrogens, genistein and daidzein, induce a decrease of methylation of BRCA1, GSTP1 and EPHB2 promoters. Therefore, soy phytoestrogens may have a protective effect on prostate cancer. However, more studies are needed in order to understand the mechanism by which genistein and daidzein have an inhibiting action on DNA methylation.
Collapse
Affiliation(s)
- Mawussi Adjakly
- Département d'Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Nagaraju GPC, Sharma D. Anti-cancer role of SPARC, an inhibitor of adipogenesis. Cancer Treat Rev 2011; 37:559-66. [PMID: 21237573 PMCID: PMC3139810 DOI: 10.1016/j.ctrv.2010.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/01/2010] [Accepted: 12/07/2010] [Indexed: 12/17/2022]
Abstract
SPARC (a secreted protein acidic and rich in cysteine) has a reputation for being potent anti-cancer and anti-obesity molecule. It is one of the first known matricellular protein that modulates interactions between cells and extracellular matrix (ECM) and is associated with the 'balance' of white adipose tissue (WAT) as well as lipogenesis and lipolysis during adipogenesis. Adipogenesis is an indication for the development of obesity and has been related to a wide variety of cancers including breast cancer, endometrial cancer, esophageal cancer, etc. Adipogenesis mainly involves ECM remodeling, changes in cell-ECM interactions, and cytoskeletal rearrangement. SPARC can also prevent hypertrophy of adipocytes and hyperplasia of adipocyte progenitors. In addition to SPARC's inhibitory role in adipogenesis, it has also been known to be involved in cell cycle, cell proliferation, cell invasion, adhesion, migration, angiogenesis and apoptosis. Molecular cancer biology and clinical biochemistry have significantly enhanced our understanding of the mechanisms that motivate the anti-cancer and anti-obesity action of SPARC. Recent studies elucidating the signaling pathways that are activated by SPARC can help develop the beneficial aspects of SPARC for cancer therapy and obesity prevention. This review focuses on the anti-cancer role of SPARC as it pertains to obesity.
Collapse
Affiliation(s)
| | - Dipali Sharma
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta GA 30322
- Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta GA 30322
| |
Collapse
|
42
|
Bonazzi VF, Nancarrow DJ, Stark MS, Moser RJ, Boyle GM, Aoude LG, Schmidt C, Hayward NK. Cross-platform array screening identifies COL1A2, THBS1, TNFRSF10D and UCHL1 as genes frequently silenced by methylation in melanoma. PLoS One 2011; 6:e26121. [PMID: 22028813 PMCID: PMC3197591 DOI: 10.1371/journal.pone.0026121] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 09/20/2011] [Indexed: 12/15/2022] Open
Abstract
Epigenetic regulation of tumor suppressor genes (TSGs) has been shown to play a central role in melanomagenesis. By integrating gene expression and methylation array analysis we identified novel candidate genes frequently methylated in melanoma. We validated the methylation status of the most promising genes using highly sensitive Sequenom Epityper assays in a large panel of melanoma cell lines and resected melanomas, and compared the findings with those from cultured melanocytes. We found transcript levels of UCHL1, COL1A2, THBS1 and TNFRSF10D were inversely correlated with promoter methylation. For THBS1 and UCHL1 the effect of this methylation on expression was confirmed at the protein level. Identification of these candidate TSGs and future research designed to understand how their silencing is related to melanoma development will increase our understanding of the etiology of this cancer and may provide tools for its early diagnosis.
Collapse
Affiliation(s)
- Vanessa F Bonazzi
- Oncogenomics Laboratory, Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Ahmad ST, Arjumand W, Seth A, Saini AK, Sultana S. Methylation of the APAF-1 and DAPK-1 promoter region correlates with progression of renal cell carcinoma in North Indian population. Tumour Biol 2011; 33:395-402. [PMID: 21922274 DOI: 10.1007/s13277-011-0235-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 08/26/2011] [Indexed: 12/31/2022] Open
Abstract
Aberrant promoter hypermethylation of cancer associated genes occur frequently during carcinogenesis and may serve as a cancer biomarker. The aim of this study was to investigate the occurrence and relevance of promoter methylation of the tumor suppressor DAPK-1, APAF-1 () and SPARC in relation to different pathological stages and histological grades of tumor progression that might act as possible independent prognostic factor in the susceptibility towards renal cell carcinoma (RCC) in North Indian population. Three tumor suppressor gene promoters namely APAF-1, DAPK-1 and SPARC were assessed by methylation-specific PCR (MS-PCR) in 196 primarily resected renal cell tumors paired with the corresponding normal tissue samples. After genomic DNA isolation and sodium bisulfite modification, methylation levels were determined and correlated with standard clinicopathological parameters, pathological stage and Fuhrman nuclear grade of RCC. Significant differences in methylation frequency among the four subtypes of renal tumors were found for APAF-1 (p < 0.001), DAPK-1 (p < 0.001) and SPARC (p = 0.182), when compared with the corresponding normal tissue. Male subjects showed stronger association of methylation frequency of all the three genes with RCC than the female subjects. Additionally, higher frequency of APAF-1, DAPK-1 and SPARC promoter methylation were directly correlated with higher tumor stage (p (trend) < 0.001). Higher frequency of promoter methylation of APAF-1 and SPARC were also associated with higher nuclear grade (p < 0.001 and p = 0.036, respectively). This gene panel might contribute to a more optimal diagnostic coverage and information, improving preoperative assessment and therapeutic decision-making in patients harboring suspicious renal masses.
Collapse
Affiliation(s)
- Shiekh Tanveer Ahmad
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Jamia Hamdard, Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | | | | | | | | |
Collapse
|
44
|
Lind GE, Danielsen SA, Ahlquist T, Merok MA, Andresen K, Skotheim RI, Hektoen M, Rognum TO, Meling GI, Hoff G, Bretthauer M, Thiis-Evensen E, Nesbakken A, Lothe RA. Identification of an epigenetic biomarker panel with high sensitivity and specificity for colorectal cancer and adenomas. Mol Cancer 2011; 10:85. [PMID: 21777459 PMCID: PMC3166273 DOI: 10.1186/1476-4598-10-85] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 07/21/2011] [Indexed: 12/16/2022] Open
Abstract
Background The presence of cancer-specific DNA methylation patterns in epithelial colorectal cells in human feces provides the prospect of a simple, non-invasive screening test for colorectal cancer and its precursor, the adenoma. This study investigates a panel of epigenetic markers for the detection of colorectal cancer and adenomas. Methods Candidate biomarkers were subjected to quantitative methylation analysis in test sets of tissue samples from colorectal cancers, adenomas, and normal colonic mucosa. All findings were verified in independent clinical validation series. A total of 523 human samples were included in the study. Receiver operating characteristic (ROC) curve analysis was used to evaluate the performance of the biomarker panel. Results Promoter hypermethylation of the genes CNRIP1, FBN1, INA, MAL, SNCA, and SPG20 was frequent in both colorectal cancers (65-94%) and adenomas (35-91%), whereas normal mucosa samples were rarely (0-5%) methylated. The combined sensitivity of at least two positives among the six markers was 94% for colorectal cancers and 93% for adenoma samples, with a specificity of 98%. The resulting areas under the ROC curve were 0.984 for cancers and 0.968 for adenomas versus normal mucosa. Conclusions The novel epigenetic marker panel shows very high sensitivity and specificity for both colorectal cancers and adenomas. Our findings suggest this biomarker panel to be highly suitable for early tumor detection.
Collapse
Affiliation(s)
- Guro E Lind
- Department of Cancer Prevention, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nocchi L, Tomasetti M, Amati M, Neuzil J, Santarelli L, Saccucci F. Thrombomodulin is silenced in malignant mesothelioma by a poly(ADP-ribose) polymerase-1-mediated epigenetic mechanism. J Biol Chem 2011; 286:19478-88. [PMID: 21489980 PMCID: PMC3103327 DOI: 10.1074/jbc.m110.217331] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 03/22/2011] [Indexed: 11/06/2022] Open
Abstract
Malignant mesothelioma (MM) is often complicated by thromboembolic episodes, with thrombomodulin (TM) playing a critical role in the anticoagulant process. Heterogeneous expression of TM has been observed in cancer, and low or no TM expression in cancer cells is associated with poor prognosis. In this study, we analyzed TM expression in biopsies of MM patients and compared them with normal mesothelial tissue. The role of DNA methylation-associated gene silencing in TM expression was investigated. To evaluate poly(ADP-ribose) polymerase-1 (PARP1) as responsible for gene promoter epigenetic modifications, nonmalignant mesothelial cells (Met-5A) and MM cells (H28) were silenced for PARP1 and the DNA methylation/acetylation-associated TM expression evaluated. A correlation between low TM expression and high level of TM promoter methylation was found in MM biopsies. Low expression of TM was restored in MM cells by their treatment with 5-aza-2'-deoxycytidine and, to a lesser extent, with trichostatin, whereas the epigenetic agents did not affect TM expression in Met-5A cells. Silencing of PARP1 resulted in a strong down-regulation of TM expression in Met-5A cells, while restoring TM expression in H28 cells. PARP1 silencing induced TM promoter methylation in Met-5A cells and demethylation in MM cells, and this was paralleled by corresponding changes in the DNA methyltransferase activity. We propose that methylation of the TM promoter is responsible for silencing of TM expression in MM tissue, a process that is regulated by PARP1.
Collapse
Affiliation(s)
- Linda Nocchi
- From the Department of Biochemistry, Biology, and Genetics, and
| | - Marco Tomasetti
- Department of Molecular Pathology and Innovative Therapies, Polytechnic University of Marche, Ancona 60126, Italy
| | - Monica Amati
- Department of Molecular Pathology and Innovative Therapies, Polytechnic University of Marche, Ancona 60126, Italy
| | - Jiri Neuzil
- the Apoptosis Research Group, School of Medical Science and Griffith Health Institute, Griffith University, Southport, Queensland 4222, Australia, and
- the Molecular Therapy Group, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Lory Santarelli
- Department of Molecular Pathology and Innovative Therapies, Polytechnic University of Marche, Ancona 60126, Italy
| | - Franca Saccucci
- From the Department of Biochemistry, Biology, and Genetics, and
| |
Collapse
|
46
|
Methylation profile of single hepatocytes derived from hepatitis B virus-related hepatocellular carcinoma. PLoS One 2011; 6:e19862. [PMID: 21625442 PMCID: PMC3100314 DOI: 10.1371/journal.pone.0019862] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 04/05/2011] [Indexed: 12/21/2022] Open
Abstract
Background With the development of high-throughput screening, a variety of genetic alterations has been found in hepatocellular carcinoma (HCC). Although previous studies on HCC methylation profiles have focused on liver tissue, studies using isolated hepatocytes are rare. The heterogeneity of liver composition may impact the genuine methylation status of HCC; therefore, it is important to clarify the methylation profile of hepatocytes to aid in understanding the process of tumorigenesis. Methods and Findings The global methylation profile of single hepatocytes isolated from liver tissue of hepatitis B virus (HBV) related HCC (HBHC) was analyzed using Illumina Infinium Human Methylation27 BeadChips, and combined bisulfite restriction analysis (COBRA) and bisulfite sequencing were used to validate the 20 significant hypermethylated genes identified. In this study, we found many noteworthy differences in the genome-wide methylation profiles of single hepatocytes of HBHC. Unsupervised hierarchical clustering analysis showed that hepatocyte methylation profiles could be classified according to three cell types: hepatocytes of HCC, adjacent hepatocytes and normal hepatocytes. Among the 20 most hypermethylated genes in the hepatocytes of HBHC, 7 novel genes (WNK2, EMILIN2, TLX3, TM6SF1, TRIM58, HIST1H4Fand GRASP) were found to be hypermethylated in HBHC and hypomethylated in paired adjacent liver tissues; these findings have not been reported in previous studies on tissue samples. Conclusion The genome-wide methylation profile of purified single hepatocytes of HBHC was aided in understanding the process of tumorigenesis, and a series of novel methylated genes found in this study have the potential to be biomarkers for the diagnosis and prognosis of HBHC.
Collapse
|
47
|
Augmented osteolysis in SPARC-deficient mice with bone-residing prostate cancer. Neoplasia 2011; 13:31-9. [PMID: 21245938 DOI: 10.1593/neo.10998] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 09/27/2010] [Accepted: 09/28/2010] [Indexed: 11/18/2022]
Abstract
Prostate cancer preferentially metastasizes to bone, which is rich in structural and matricellular proteins capable of altering prostate cancer progression. This study explores the role of the bone stromal matricellular protein SPARC (osteonectin/BM-40) in the progression of bone metastatic prostate cancer. Quantification of bone destruction analyzed by micro-computed tomography showed augmented osteoclastic resorption, characterized by decreases in several morphometric bone parameters in SPARC knock out (KO) tibiae harboring RM1 murine prostate cancer cells compared with wild type (WT) animals. Tumor progression stimulated osteoclast formation, which was augmented in SPARC KO mice. In vitro differentiation of SPARC KO osteoclasts indicated accelerated progenitor expansion and formation of tartrate-resistant acid phosphatase-positive osteoclast-like cells with increased resorptive capacity, a mechanism resulting in enhanced tumor-induced bone loss in vivo. Whereas altered bone structure due to SPARC KO played a role in increased osteolysis, the enhanced osteolysis was primarily the result of increased resorption by SPARC KO osteoclasts. Our findings indicate that bone stromal SPARC suppresses tumor-induced bone lesion expansion by limiting osteoclast maturation and function.
Collapse
|
48
|
Epigenetic regulation of cell life and death decisions and deregulation in cancer. Essays Biochem 2010; 48:121-46. [PMID: 20822491 DOI: 10.1042/bse0480121] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For every cell, there is a time to live and a time to die. It is apparent that cell life and death decisions are taken by individual cells based on their interpretation of physiological or non-physiological stimuli, or their own self-assessment of internal damage or changes in their environment. Apoptosis or programmed cell death is a key regulator of physiological growth control and regulation of tissue homoeostasis. One of the most important advances in cancer research in recent years is the recognition that cell death, mostly by apoptosis, is crucially involved in the regulation of tumour formation and also critically determines treatment response. The initiation and progression of cancer, traditionally seen as a genetic disease, is now realized to involve epigenetic abnormalities along with genetic alterations. The study of epigenetic mechanisms in cancer, such as DNA methylation, histone modifications and microRNA expression, has revealed a plethora of events that contribute to the neoplastic phenotype through stable changes in the expression of genes critical to cell death pathways. A better understanding of the epigenetic molecular events that regulate apoptosis, together with the reversible nature of epigenetic aberrations, should contribute to the emergence of the promising field of epigenetic therapy.
Collapse
|
49
|
Portela M, Casas-Tinto S, Rhiner C, López-Gay JM, Domínguez O, Soldini D, Moreno E. Drosophila SPARC Is a Self-Protective Signal Expressed by Loser Cells during Cell Competition. Dev Cell 2010; 19:562-73. [DOI: 10.1016/j.devcel.2010.09.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 08/03/2010] [Accepted: 08/17/2010] [Indexed: 12/26/2022]
|
50
|
Berretta R, Moscato P. Cancer biomarker discovery: the entropic hallmark. PLoS One 2010; 5:e12262. [PMID: 20805891 PMCID: PMC2923618 DOI: 10.1371/journal.pone.0012262] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 06/26/2010] [Indexed: 12/29/2022] Open
Abstract
Background It is a commonly accepted belief that cancer cells modify their transcriptional state during the progression of the disease. We propose that the progression of cancer cells towards malignant phenotypes can be efficiently tracked using high-throughput technologies that follow the gradual changes observed in the gene expression profiles by employing Shannon's mathematical theory of communication. Methods based on Information Theory can then quantify the divergence of cancer cells' transcriptional profiles from those of normally appearing cells of the originating tissues. The relevance of the proposed methods can be evaluated using microarray datasets available in the public domain but the method is in principle applicable to other high-throughput methods. Methodology/Principal Findings Using melanoma and prostate cancer datasets we illustrate how it is possible to employ Shannon Entropy and the Jensen-Shannon divergence to trace the transcriptional changes progression of the disease. We establish how the variations of these two measures correlate with established biomarkers of cancer progression. The Information Theory measures allow us to identify novel biomarkers for both progressive and relatively more sudden transcriptional changes leading to malignant phenotypes. At the same time, the methodology was able to validate a large number of genes and processes that seem to be implicated in the progression of melanoma and prostate cancer. Conclusions/Significance We thus present a quantitative guiding rule, a new unifying hallmark of cancer: the cancer cell's transcriptome changes lead to measurable observed transitions of Normalized Shannon Entropy values (as measured by high-througput technologies). At the same time, tumor cells increment their divergence from the normal tissue profile increasing their disorder via creation of states that we might not directly measure. This unifying hallmark allows, via the the Jensen-Shannon divergence, to identify the arrow of time of the processes from the gene expression profiles, and helps to map the phenotypical and molecular hallmarks of specific cancer subtypes. The deep mathematical basis of the approach allows us to suggest that this principle is, hopefully, of general applicability for other diseases.
Collapse
Affiliation(s)
- Regina Berretta
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Information Based Medicine Program, Hunter Medical Research Institute, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Pablo Moscato
- Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, The University of Newcastle, Callaghan, New South Wales, Australia
- Information Based Medicine Program, Hunter Medical Research Institute, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
- Australian Research Council Centre of Excellence in Bioinformatics, Callaghan, New South Wales, Australia
- * E-mail:
| |
Collapse
|