1
|
Wang L, Zhang J, Xia M, Liu C, Zu X, Zhong J. High Mobility Group A1 (HMGA1): Structure, Biological Function, and Therapeutic Potential. Int J Biol Sci 2022; 18:4414-4431. [PMID: 35864955 PMCID: PMC9295051 DOI: 10.7150/ijbs.72952] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein characterized by no transcriptional activity. It mainly plays a regulatory role by modifying the structure of DNA. A large number of studies have confirmed that HMGA1 regulates genes related to tumours in the reproductive system, digestive system, urinary system and haematopoietic system. HMGA1 is rare in adult cells and increases in highly proliferative cells such as embryos. After being stimulated by external factors, it will produce effects through the Wnt/β-catenin, PI3K/Akt, Hippo and MEK/ERK pathways. In addition, HMGA1 also affects the ageing, apoptosis, autophagy and chemotherapy resistance of cancer cells, which are linked to tumorigenesis. In this review, we summarize the mechanisms of HMGA1 in cancer progression and discuss the potential clinical application of targeted HMGA1 therapy, indicating that targeted HMGA1 is of great significance in the diagnosis and treatment of malignancy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Min Xia
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Chang Liu
- Department of Endocrinology and Metabolism, The First People's Hospital of Chenzhou, First School of Clinical Medicine, University of Southern Medical, Guangzhou 510515, Guangdong, China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| |
Collapse
|
2
|
Pegoraro S, Ros G, Sgubin M, Petrosino S, Zambelli A, Sgarra R, Manfioletti G. Targeting the intrinsically disordered architectural High Mobility Group A (HMGA) oncoproteins in breast cancer: learning from the past to design future strategies. Expert Opin Ther Targets 2020; 24:953-969. [PMID: 32970506 DOI: 10.1080/14728222.2020.1814738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat because of its heterogeneity and lack of specific therapeutic targets. High Mobility Group A (HMGA) proteins are chromatin architectural factors that have multiple oncogenic functions in breast cancer, and they represent promising molecular therapeutic targets for this disease. AREAS COVERED We offer an overview of the strategies that have been exploited to counteract HMGA oncoprotein activities at the transcriptional and post-transcriptional levels. We also present the possibility of targeting cancer-associated factors that lie downstream of HMGA proteins and discuss the contribution of HMGA proteins to chemoresistance. EXPERT OPINION Different strategies have been exploited to counteract HMGA protein activities; these involve interfering with their nucleic acid binding properties and the blocking of HMGA expression. Some approaches have provided promising results. However, some unique characteristics of the HMGA proteins have not been exploited; these include their extensive protein-protein interaction network and their intrinsically disordered status that present the possibility that HMGA proteins could be involved in the formation of proteinaceous membrane-less organelles (PMLO) by liquid-liquid phase separation. These unexplored characteristics could open new pharmacological avenues to counteract the oncogenic contributions of HMGA proteins.
Collapse
Affiliation(s)
- Silvia Pegoraro
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Gloria Ros
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Michela Sgubin
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Sara Petrosino
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | |
Collapse
|
3
|
Lamb LS, Sim HW, McCormack AI. Exploring the Role of Novel Medical Therapies for Aggressive Pituitary Tumors: A Review of the Literature-"Are We There Yet?". Cancers (Basel) 2020; 12:cancers12020308. [PMID: 32012988 PMCID: PMC7072681 DOI: 10.3390/cancers12020308] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
Aggressive pituitary tumors account for up to 10% of pituitary tumors and are characterized by resistance to medical treatment and multiple recurrences despite standard therapies, including surgery, radiotherapy, and chemotherapy. They are associated with increased morbidity and mortality, particularly pituitary carcinomas, which have mortality rates of up to 66% at 1 year after diagnosis. Novel targeted therapies under investigation include mammalian target of rapamycin (mTOR), tyrosine kinase, and vascular endothelial growth factor (VEGF) inhibitors. More recently, immune checkpoint inhibitors have been proposed as a potential treatment option for pituitary tumors. An increased understanding of the molecular pathogenesis of aggressive pituitary tumors is required to identify potential biomarkers and therapeutic targets. This review discusses novel approaches to the management of aggressive pituitary tumors and the role of molecular profiling.
Collapse
Affiliation(s)
- Lydia S. Lamb
- Department of Endocrinology, St Vincent’s Hospital, Sydney, NSW 2010, Australia;
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
| | - Hao-Wen Sim
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
- Kinghorn Cancer Centre, Sydney, NSW 2010, Australia
| | - Ann I. McCormack
- Department of Endocrinology, St Vincent’s Hospital, Sydney, NSW 2010, Australia;
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
- Correspondence: ; Tel.: +61-2-9295-8489
| |
Collapse
|
4
|
Belaya Z, Khandaeva P, Nonn L, Nikitin A, Solodovnikov A, Sitkin I, Grigoriev A, Pikunov M, Lapshina A, Rozhinskaya L, Melnichenko G, Dedov I. Circulating Plasma microRNA to Differentiate Cushing's Disease From Ectopic ACTH Syndrome. Front Endocrinol (Lausanne) 2020; 11:331. [PMID: 32582027 PMCID: PMC7291947 DOI: 10.3389/fendo.2020.00331] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Corticotropinomas and adrenocorticotropic hormone (ACTH)-secreting neuroendocrine tumors exhibit differential levels of some microRNAs (miRs) compared to normal tissue. Because miRs can be released from tissues into circulation, they offer promise as novel disease biomarkers. Objective: To evaluate whether miRs are differentially detected in plasma samples of patients with ACTH-dependent Cushing's syndrome (CS). Design: Case-control study. Methods: Morning fasting plasma samples were collected from 41 consecutive patients with confirmed ACTH-dependent CS and 11 healthy subjects and stored at -80°C. Twenty-one miRs previously reported to be differentially expressed in ACTH-secreting tumors vs. healthy tissue samples were quantified in plasma by qPCR. Results: Among enrolled subjects, 28 were confirmed to have Cushing's disease (CD), 13 had ectopic ACTH secretion (EAS) and 11 were healthy controls. We found statistically significant differences in the circulating levels of miR-16-5p [45.04 (95% CI 28.77-61.31) in CD vs. 5.26 (2.65-7.87) in EAS, P < 0.001; q = 0.001], miR-145-5p [0.097 (0.027-0.167) in CD vs. undetectable levels in EAS, P = 0.008; q = 0.087] and differences in miR-7g-5p [1.842 (1.283-2.400) in CD vs. 0.847 (0.187-1.507) in EAS, P = 0.02; q = 0.14]. The area under the receiver-operator (ROC) curve was 0.879 (95% CI 0.770-0.987), p < 0.001, when using miR-16-5p to distinguish between CD and EAS. Circulating levels of miR-16-5p in the healthy control group differed from that of both the CD and EAS groups. Conclusions: Plasma miR levels differ in patients with CD and EAS. In particular, miR-16-5p, miR-145-5p and miR-7g-5p are promising biomarkers for further research to differentiate ACTH-dependent CS.
Collapse
Affiliation(s)
- Zhanna Belaya
- The National Medical Research Centre for Endocrinology, Moscow, Russia
- *Correspondence: Zhanna Belaya
| | - Patimat Khandaeva
- The National Medical Research Centre for Endocrinology, Moscow, Russia
| | - Larisa Nonn
- Department Pathology College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Alexey Nikitin
- Federal Research and Clinical Center FMBA of Russia, Moscow, Russia
| | | | - Ivan Sitkin
- The National Medical Research Centre for Endocrinology, Moscow, Russia
| | - Andrey Grigoriev
- The National Medical Research Centre for Endocrinology, Moscow, Russia
| | - Mikhail Pikunov
- National Medical Research Center of Surgery Named After A.V. Vishnevsky, Moscow, Russia
| | | | | | | | - Ivan Dedov
- The National Medical Research Centre for Endocrinology, Moscow, Russia
| |
Collapse
|
5
|
Kober P, Boresowicz J, Rusetska N, Maksymowicz M, Paziewska A, Dąbrowska M, Kunicki J, Bonicki W, Ostrowski J, Siedlecki JA, Bujko M. The Role of Aberrant DNA Methylation in Misregulation of Gene Expression in Gonadotroph Nonfunctioning Pituitary Tumors. Cancers (Basel) 2019; 11:E1650. [PMID: 31731486 PMCID: PMC6895980 DOI: 10.3390/cancers11111650] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Gonadotroph nonfunctioning pituitary adenomas (NFPAs) are common intracranial tumors, but the role of aberrant epigenetic regulation in their development remains poorly understood. In this study, we investigated the effect of impaired CpG methylation in NFPAs. We determined DNA methylation and transcriptomic profiles in 32 NFPAs and normal pituitary sections using methylation arrays and sequencing, respectively. Ten percent of differentially methylated CpGs were correlated with gene expression, and the affected genes are involved in a variety of tumorigenesis-related pathways. Different proportions of gene body and promoter region localization were observed in CpGs with negative and positive correlations between methylation and gene expression, and different proportions of CpGs were located in 'open sea' and 'shelf/shore' regions. The expression of ~8% of genes differentially expressed in NFPAs was related to aberrant methylation. Methylation levels of seven CpGs located in the regulatory regions of FAM163A, HIF3A and PRSS8 were determined by pyrosequencing, and gene expression was measured by qRT-PCR and immunohistochemistry in 83 independent NFPAs. The results clearly confirmed the negative correlation between methylation and gene expression for these genes. By identifying which aberrantly methylated CpGs affect gene expression in gonadotrophinomas, our data confirm the role of aberrant methylation in pathogenesis of gonadotroph NFPAs.
Collapse
Affiliation(s)
- Paulina Kober
- Department of Molecular and Translational Oncology, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (P.K.); (J.B.); (N.R.); (J.A.S.)
| | - Joanna Boresowicz
- Department of Molecular and Translational Oncology, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (P.K.); (J.B.); (N.R.); (J.A.S.)
| | - Natalia Rusetska
- Department of Molecular and Translational Oncology, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (P.K.); (J.B.); (N.R.); (J.A.S.)
| | - Maria Maksymowicz
- Department of Pathology and Laboratory Diagnostics, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland;
| | - Agnieszka Paziewska
- Department of Genetics, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland
| | - Michalina Dąbrowska
- Department of Genetics, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
| | - Jacek Kunicki
- Department of Neurosurgery, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (J.K.); (W.B.)
| | - Wiesław Bonicki
- Department of Neurosurgery, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (J.K.); (W.B.)
| | - Jerzy Ostrowski
- Department of Genetics, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (A.P.); (M.D.); (J.O.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland
| | - Janusz A. Siedlecki
- Department of Molecular and Translational Oncology, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (P.K.); (J.B.); (N.R.); (J.A.S.)
| | - Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Skłodowska-Curie Institute—Oncology Center, 02-034 Warsaw, Poland; (P.K.); (J.B.); (N.R.); (J.A.S.)
| |
Collapse
|
6
|
Hauser BM, Lau A, Gupta S, Bi WL, Dunn IF. The Epigenomics of Pituitary Adenoma. Front Endocrinol (Lausanne) 2019; 10:290. [PMID: 31139150 PMCID: PMC6527758 DOI: 10.3389/fendo.2019.00290] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background: The vast majority of pituitary tumors are benign and behave accordingly; however, a fraction are invasive and are more aggressive, with a very small fraction being frankly malignant. The cellular pathways that drive transformation in pituitary neoplasms are poorly characterized, and current classification methods are not reliable correlates of clinical behavior. Novel techniques in epigenetics, the study of alterations in gene expression without changes to the genetic code, provide a new dimension to characterize tumors, and may hold implications for prognostication and management. Methods: We conducted a review of primary epigenetic studies of pituitary tumors with a focus on histone modification, DNA methylation, and transcript modification. Results: High levels of methylation have been identified in invasive and large pituitary tumors. DNA methyltransferase overexpression has been detected in pituitary tumors, especially in macroadenomas. Methylation differences at CpG sites in promoter regions may distinguish several types of tumors from normal pituitary tissue. Histone modifications have been linked to increased p53 expression and longer progression-free survival in pituitary tumors; sirtuins are expressed at higher values in GH-expressing compared to nonfunctional adenomas and correlate inversely with size in somatotrophs. Upregulation in citrullinating enzymes may be an early pathogenic marker of prolactinomas. Numerous genes involved with cell growth and signaling show altered methylation status for pituitary tumors, including cell cycle regulators, components of signal transduction pathways, apoptotic regulators, and pituitary developmental signals. Conclusions: The limited clinical predictive capacity of the current pituitary tumor classification system suggests that tumor subclasses likely remain to be discovered. Ongoing epigenetic studies could provide a basis for adding methylation and/or acetylation screening to standard pituitary tumor workups. Identifying robust correlations between tumor epigenetics and corresponding histological, radiographic, and clinical course information could ultimately inform clinical decision-making.
Collapse
Affiliation(s)
- Blake M. Hauser
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ashley Lau
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Saksham Gupta
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Wenya Linda Bi
| | - Ian F. Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Ian F. Dunn
| |
Collapse
|
7
|
Histone Citrullination Represses MicroRNA Expression, Resulting in Increased Oncogene mRNAs in Somatolactotrope Cells. Mol Cell Biol 2018; 38:MCB.00084-18. [PMID: 29987187 DOI: 10.1128/mcb.00084-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/29/2018] [Indexed: 12/12/2022] Open
Abstract
Peptidylarginine deiminase (PAD) enzymes convert histone arginine residues into citrulline to modulate chromatin organization and gene expression. Although PADs are expressed in anterior pituitary gland cells, their functional role and expression in pituitary adenomas are unknown. To begin to address these issues, we first examined normal human pituitaries and pituitary adenomas and found that PAD2, PAD4, and citrullinated histones are highest in prolactinomas and somatoprolactinomas. In the somatoprolactinoma-derived GH3 cell line, PADs citrullinate histone H3, which is attenuated by a pan-PAD inhibitor. RNA sequencing and chromatin immunoprecipitation (ChIP) studies show that the expression of microRNAs (miRNAs) let-7c-2, 23b, and 29c is suppressed by histone citrullination. Our studies demonstrate that these miRNAs directly target the mRNA of the oncogenes encoding HMGA, insulin-like growth factor 1 (IGF-1), and N-MYC, which are highly implicated in human prolactinoma/somatoprolactinoma pathogenesis. Our results are the first to define a direct role for PAD-catalyzed histone citrullination in miRNA expression, which may underlie the etiology of prolactinoma and somatoprolactinoma tumors through regulation of oncogene expression.
Collapse
|
8
|
Ezzat S, Cheng S, Asa SL. Epigenetics of pituitary tumors: Pathogenetic and therapeutic implications. Mol Cell Endocrinol 2018; 469:70-76. [PMID: 28711607 DOI: 10.1016/j.mce.2017.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/21/2017] [Accepted: 07/11/2017] [Indexed: 11/25/2022]
Abstract
Genetic mutations involving oncogenes or tumor suppressor genes are relatively uncommon in human sporadic pituitary tumors. Instead, increasing evidence has highlighted frequent epigenetic alterations including DNA methylation, histone modifications, and enhanced miRNA expression. This review covers some of this evidence as it illuminates mechanisms of tumorigenesis and highlights therapeutic opportunities.
Collapse
Affiliation(s)
- Shereen Ezzat
- Department of Medicine, University of Toronto, The Endocrine Oncology Site Group, Princess Margaret Cancer Centre, and The Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
| | - Sonia Cheng
- Department of Medicine, University of Toronto, The Endocrine Oncology Site Group, Princess Margaret Cancer Centre, and The Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Sylvia L Asa
- Department of Laboratory Medicine & Pathobiology, University of Toronto, The Endocrine Oncology Site Group, Princess Margaret Cancer Centre, and The Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Wierinckx A, Roche M, Legras-Lachuer C, Trouillas J, Raverot G, Lachuer J. MicroRNAs in pituitary tumors. Mol Cell Endocrinol 2017; 456:51-61. [PMID: 28089822 DOI: 10.1016/j.mce.2017.01.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/14/2016] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Since the presence of microRNAs was first observed in normal pituitary, the majority of scientific publications addressing their role and the function of microRNAs in the pituitary have been based on pituitary tumor studies. In this review, we briefly describe the involvement of microRNAs in the synthesis of pituitary hormones and we present a comprehensive inventory of microRNA suppressors and inducers of pituitary tumors. Finally, we summarize the functional role of microRNAs in tumorigenesis, progression and aggressiveness of pituitary tumors, mechanisms contributing to the regulation (transcription factors, genomic modifications or epigenetic) or modulation (pharmacological treatment) of microRNAs in these tumors, and the interest of thoroughly studying the expression of miRNAs in body fluids.
Collapse
Affiliation(s)
- Anne Wierinckx
- Université Lyon 1, Université de Lyon, Lyon, France; Institut Universitaire de Technologie Lyon1, Université de Lyon, F-69622 Villeurbanne Cedex, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France.
| | | | - Catherine Legras-Lachuer
- Université Lyon 1, Université de Lyon, Lyon, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France; ViroScan3D, F-01600 Trévoux, France; UMR CNRS 5557 UCBL USC INRA 1193 ENVL, Dynamique Microbienne et Transmission Virale, F-69100 Villeurbanne Cedex, France
| | - Jacqueline Trouillas
- Université Lyon 1, Université de Lyon, Lyon, France; Centre de Pathologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron F-69677, France
| | - Gérald Raverot
- Université Lyon 1, Université de Lyon, Lyon, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; Fédération d'Endocrinologie, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, F-69677, France Université Lyon 1, Université de Lyon, Lyon, France
| | - Joël Lachuer
- Université Lyon 1, Université de Lyon, Lyon, France; Institut Universitaire de Technologie Lyon1, Université de Lyon, F-69622 Villeurbanne Cedex, France; INSERM U1052, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; CNRS UMR 5286, Cancer Research Center of Lyon, F-69373 Lyon Cedex 08, France; ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, F-69373 Lyon Cedex 08, France
| |
Collapse
|
10
|
Biersack B. Non-coding RNA/microRNA-modulatory dietary factors and natural products for improved cancer therapy and prevention: Alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins. Noncoding RNA Res 2016; 1:51-63. [PMID: 30159411 PMCID: PMC6096427 DOI: 10.1016/j.ncrna.2016.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023] Open
Abstract
Non-coding small RNA molecules, the microRNAs (miRNAs), contribute decisively to the epigenetic regulation processes in cancer cells. Problematic pathogenic properties of cancer cells and the response of cancers towards anticancer drugs are highly influenced by miRNAs. Both increased drug activity and formation of tumor resistance are regulated by miRNAs. Further to this, the survival and proliferation of cancer cells and the formation of metastases is based on the modulated expression of certain miRNAs. In particular, drug-resistant cancer stem-like cells (CSCs) depend on the presence and absence of specific miRNAs. Fortunately, several small molecule natural compounds were discovered that target miRNAs involved in the modulation of tumor aggressiveness and drug resistance. This review gives an overview of the effects of a selection of naturally occurring small molecules (alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins) on miRNAs that are closely tangled with cancer diseases.
Collapse
Key Words
- AM, allyl mercaptan
- AOM, azoxymethane
- Aliphatic carboxylic acids
- Alkaloids
- Anticancer drugs
- CPT, camptothecin
- DADS, diallyl disulfide
- DHA, docosahexaenoic acid
- DIM, 3,3′-diindolylmethane
- EPA, eicosapentaenoic acid
- FA, folic acid
- GTC, green tea catechins
- I3C, indole-3-carbinol
- MiRNA
- NaB, sodium butyrate
- Organosulfur compounds
- PEITC, phenethylisothiocyanate
- PUFA, polyunsaturated fatty acid
- SAMC, S-allylmercaptocysteine
- SFN, sulforaphane
- TSA, trichostatin A
- Water-soluble vitamins
Collapse
|
11
|
Abstract
MicroRNAs (miRNAs) are non-coding RNAs generated from endogenous hairpin-shaped transcripts that powerfully regulate gene expression at post-transcriptional level. Each miRNA is capable to regulate the expression levels of hundreds of transcripts and each mRNA may have more than one miRNA recognition sequence. There is emerging evidence that deregulation of miRNA expression leads to the alteration of pivotal physiological functions contributing to the development of diseases and neoplasms, including pituitary adenoma. This review is aimed at providing the up-to-date knowledge concerning deregulated miRNAs of pituitary tumors and their functions. In order to take stock, pituitary tumors have been sub-divided in different classes on the basis of tumor features (histotype, dimension, aggressiveness). The overview takes full consideration of the recent advances in miRNAs role as potential therapeutics and biomarkers.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Ettore Degli Uberti
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy.
| |
Collapse
|
12
|
Kitchen MO, Bryan RT, Emes RD, Glossop JR, Luscombe C, Cheng KK, Zeegers MP, James ND, Devall AJ, Mein CA, Gommersall L, Fryer AA, Farrell WE. Quantitative genome-wide methylation analysis of high-grade non-muscle invasive bladder cancer. Epigenetics 2016; 11:237-46. [PMID: 26929985 DOI: 10.1080/15592294.2016.1154246] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
High-grade non-muscle invasive bladder cancer (HG-NMIBC) is a clinically unpredictable disease with greater risks of recurrence and progression relative to their low-intermediate-grade counterparts. The molecular events, including those affecting the epigenome, that characterize this disease entity in the context of tumor development, recurrence, and progression, are incompletely understood. We therefore interrogated genome-wide DNA methylation using HumanMethylation450 BeadChip arrays in 21 primary HG-NMIBC tumors relative to normal bladder controls. Using strict inclusion-exclusion criteria we identified 1,057 hypermethylated CpGs within gene promoter-associated CpG islands, representing 256 genes. We validated the array data by bisulphite pyrosequencing and examined 25 array-identified candidate genes in an independent cohort of 30 HG-NMIBC and 18 low-intermediate-grade NMIBC. These analyses revealed significantly higher methylation frequencies in high-grade tumors relative to low-intermediate-grade tumors for the ATP5G2, IRX1 and VAX2 genes (P<0.05), and similarly significant increases in mean levels of methylation in high-grade tumors for the ATP5G2, VAX2, INSRR, PRDM14, VSX1, TFAP2b, PRRX1, and HIST1H4F genes (P<0.05). Although inappropriate promoter methylation was not invariantly associated with reduced transcript expression, a significant association was apparent for the ARHGEF4, PON3, STAT5a, and VAX2 gene transcripts (P<0.05). Herein, we present the first genome-wide DNA methylation analysis in a unique HG-NMIBC cohort, showing extensive and discrete methylation changes relative to normal bladder and low-intermediate-grade tumors. The genes we identified hold significant potential as targets for novel therapeutic intervention either alone, or in combination, with more conventional therapeutic options in the treatment of this clinically unpredictable disease.
Collapse
Affiliation(s)
- Mark O Kitchen
- a Institute for Science and Technology in Medicine, Keele University , UK.,b Urology Department , University Hospitals of North Midlands NHS Trust , UK
| | - Richard T Bryan
- c Institute of Cancer and Genomic Sciences, University of Birmingham , UK
| | - Richard D Emes
- d Advanced Data Analysis Center, University of Nottingham , UK
| | - John R Glossop
- a Institute for Science and Technology in Medicine, Keele University , UK
| | | | - K K Cheng
- c Institute of Cancer and Genomic Sciences, University of Birmingham , UK
| | - Maurice P Zeegers
- c Institute of Cancer and Genomic Sciences, University of Birmingham , UK.,e Department of Complex Genetics , Maastricht University Medical Center , The Netherlands.,f NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , The Netherlands.,g CAPHRI School for Public Health and Primary Care, Maastricht University Medical Center , The Netherlands
| | | | - Adam J Devall
- c Institute of Cancer and Genomic Sciences, University of Birmingham , UK
| | - Charles A Mein
- i The Genome Center, Barts and the London School of Medicine and Dentistry , London , UK
| | - Lyndon Gommersall
- b Urology Department , University Hospitals of North Midlands NHS Trust , UK
| | - Anthony A Fryer
- a Institute for Science and Technology in Medicine, Keele University , UK
| | - William E Farrell
- a Institute for Science and Technology in Medicine, Keele University , UK
| |
Collapse
|
13
|
Schult D, Hölsken A, Siegel S, Buchfelder M, Fahlbusch R, Kreitschmann-Andermahr I, Buslei R. EZH2 is highly expressed in pituitary adenomas and associated with proliferation. Sci Rep 2015; 5:16965. [PMID: 26593398 PMCID: PMC4655333 DOI: 10.1038/srep16965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a core epigenetic regulator, playing a crucial role in cell cycle regulation. The protein is known to be associated with proliferation and worse outcome in several tumor entities. In this study, we immunohistochemically investigated the expression pattern of EZH2 in a large cohort of pituitary tumors. These results were correlated with clinical features and double immunofluorescence stainings (DIS) were conducted to evaluate co-expression of EZH2 and proliferation marker Ki-67. Furthermore, we analyzed the effect of EZH2 inhibition on cell proliferation in vitro using the pituitary cell line AtT-20. While in the normal anterior pituitary EZH2 was almost absent, the cohort of tumors showed enhanced expression levels (p ≤ 0.0005). This was positively associated with Ki-67 indices (r = 0.834, p ≤ 0.0005) and DIF confirmed a predominant co-expression of both markers. In vitro experiments revealed a significant (p ≤ 0.05) decrease of tumor cell proliferation using the EZH2 inhibitor GSK126. Our results further support that epigenetic events are involved in the pathogenesis and biology of pituitary adenomas (PA). Therefore, EZH2 may function as a new potential target for therapeutic interventions in PA.
Collapse
Affiliation(s)
- David Schult
- Institute of Neuropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen
| | - Annett Hölsken
- Institute of Neuropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen
| | - Sonja Siegel
- Department of Neurosurgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen.,Department of Neurosurgery, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen
| | - Rudolf Fahlbusch
- Department of Neurosurgery, International Neuroscience Institute, Rudolf-Pichlmayr-Straße 4, 30625 Hannover
| | - Ilonka Kreitschmann-Andermahr
- Department of Neurosurgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen.,Department of Neurosurgery, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen
| | - Rolf Buslei
- Institute of Neuropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen
| |
Collapse
|
14
|
Ali SR, Humphreys KJ, McKinnon RA, Michael MZ. Impact of Histone Deacetylase Inhibitors on microRNA Expression and Cancer Therapy: A Review. Drug Dev Res 2015; 76:296-317. [PMID: 26303212 DOI: 10.1002/ddr.21268] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chromatin-modifying drugs, such as histone deacetylase inhibitors (HDACi), have shown potential as cancer therapeutics, either alone or in combination with other therapies. HDACi have the ability to reverse aberrant epigenetic modifications associated with cancer, namely dysregulated histone acetylation. There are currently three FDA approved HDACi; vorinostat, romidepsin, and panobinostat. Epigenetic modifications can regulate the expression of protein coding genes, and in addition can alter expression of microRNA (miRNA) genes. Many miRNAs play key roles in cell proliferation and apoptosis, and are commonly dysregulated in cancer states. A number of in vitro and in vivo studies have demonstrated the ability of chromatin-modifying drugs to alter miRNA expression, which may provide the basis for further investigation of miRNAs as therapeutic targets or as biomarkers of drug response. This review summarises findings from studies investigating the effects of HDACi on miRNA expression, as well as key clinical trials involving HDACi. Understanding how chromatin-modifying drugs epigenetically modulate miRNA genes provides further insight into the cellular mechanisms that deliver therapeutic responses, and may assist in refining treatment strategies.
Collapse
Affiliation(s)
- Saira R Ali
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Karen J Humphreys
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia.,Department of Gastroenterology and Hepatology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|