1
|
Fontana D, Zambrotta GPM, Scannella A, Piazza R, Gambacorti-Passerini C. Late relapse of chronic myeloid leukemia after allogeneic bone marrow transplantation points to KANSARL (KANSL1::ARL17A) alteration: a case report with insights on the molecular landscape. Ann Hematol 2024; 103:1561-1568. [PMID: 38321229 PMCID: PMC11009776 DOI: 10.1007/s00277-024-05649-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
Chronic myeloid leukemia is a myeloproliferative neoplasm characterized by the presence of the Philadelphia chromosome and the consequent BCR::ABL1 oncoprotein. In the era before the introduction of tyrosine kinase inhibitors (TKIs), the only potentially curative treatment was allogeneic hematopoietic stem cell transplantation (HSCT). Here, we present the case of a patient affected by CML who experienced a relapse 20 years after allogeneic HSCT. Following relapse, the patient was treated with imatinib and bosutinib, resulting in a deep molecular response and successfully discontinued treatment. Additional analysis including whole-exome sequencing and RNA sequencing provided some insights on the molecular mechanisms of the relapse: the identification of the fusion transcript KANSL1::ARL17A (KANSARL), a cancer predisposition fusion gene, could justify a condition of genomic instability which may be associated with the onset and/or probably the late relapse of his CML.
Collapse
Affiliation(s)
- Diletta Fontana
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, Monza, MB, 20900, Italy.
| | - Giovanni Paolo Maria Zambrotta
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, Monza, MB, 20900, Italy
- Hematology Division and Bone Marrow Unit, IRCCS, San Gerardo dei Tintori, Monza, Italy
| | - Antonio Scannella
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, Monza, MB, 20900, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, Monza, MB, 20900, Italy
- Hematology Division and Bone Marrow Unit, IRCCS, San Gerardo dei Tintori, Monza, Italy
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, Monza, MB, 20900, Italy
- Hematology Division and Bone Marrow Unit, IRCCS, San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
2
|
Ferreira A, Castanheira P, Escrevente C, Barral DC, Barona T. Membrane trafficking alterations in breast cancer progression. Front Cell Dev Biol 2024; 12:1350097. [PMID: 38533085 PMCID: PMC10963426 DOI: 10.3389/fcell.2024.1350097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/12/2024] [Indexed: 03/28/2024] Open
Abstract
Breast cancer (BC) is the most common type of cancer in women, and remains one of the major causes of death in women worldwide. It is now well established that alterations in membrane trafficking are implicated in BC progression. Indeed, membrane trafficking pathways regulate BC cell proliferation, migration, invasion, and metastasis. The 22 members of the ADP-ribosylation factor (ARF) and the >60 members of the rat sarcoma (RAS)-related in brain (RAB) families of small GTP-binding proteins (GTPases), which belong to the RAS superfamily, are master regulators of membrane trafficking pathways. ARF-like (ARL) subfamily members are involved in various processes, including vesicle budding and cargo selection. Moreover, ARFs regulate cytoskeleton organization and signal transduction. RABs are key regulators of all steps of membrane trafficking. Interestingly, the activity and/or expression of some of these proteins is found dysregulated in BC. Here, we review how the processes regulated by ARFs and RABs are subverted in BC, including secretion/exocytosis, endocytosis/recycling, autophagy/lysosome trafficking, cytoskeleton dynamics, integrin-mediated signaling, among others. Thus, we provide a comprehensive overview of the roles played by ARF and RAB family members, as well as their regulators in BC progression, aiming to lay the foundation for future research in this field. This research should focus on further dissecting the molecular mechanisms regulated by ARFs and RABs that are subverted in BC, and exploring their use as therapeutic targets or prognostic markers.
Collapse
|
3
|
Malik S, Pradeep SP, Kumar V, Xiao Y, Deng Y, Fan R, Vasquez JC, Singh V, Bahal R. Antitumor efficacy of a sequence-specific DNA-targeted γPNA-based c-Myc inhibitor. Cell Rep Med 2024; 5:101354. [PMID: 38183981 PMCID: PMC10829792 DOI: 10.1016/j.xcrm.2023.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 09/21/2023] [Accepted: 12/11/2023] [Indexed: 01/08/2024]
Abstract
Targeting oncogenes at the genomic DNA level can open new avenues for precision medicine. Significant efforts are ongoing to target oncogenes using RNA-targeted and protein-targeted platforms, but no progress has been made to target genomic DNA for cancer therapy. Here, we introduce a gamma peptide nucleic acid (γPNA)-based genomic DNA-targeted platform to silence oncogenes in vivo. γPNAs efficiently invade the mixed sequences of genomic DNA with high affinity and specificity. As a proof of concept, we establish that γPNA can inhibit c-Myc transcription in multiple cell lines. We evaluate the in vivo efficacy and safety of genomic DNA targeting in three pre-clinical models. We also establish that anti-transcription γPNA in combination with histone deacetylase inhibitors and chemotherapeutic drugs results in robust antitumor activity in cell-line- and patient-derived xenografts. Overall, this strategy offers a unique therapeutic platform to target genomic DNA to inhibit oncogenes for cancer therapy.
Collapse
Affiliation(s)
- Shipra Malik
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Sai Pallavi Pradeep
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Yong Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA; Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Juan C Vasquez
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Vijender Singh
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
4
|
Bisht S, Chawla B, Kumar A, Vijayan V, Kumar M, Sharma P, Dada R. Identification of novel genes by targeted exome sequencing in Retinoblastoma. Ophthalmic Genet 2022; 43:771-788. [PMID: 35930312 DOI: 10.1080/13816810.2022.2106497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Retinoblastoma (RB) is initiated by mutation in both alleles of RB1 gene. However, few cases may occur even in the absence of RB1 mutation suggesting the role of genes other than RB1. METHODOLOGY The current study was planned to utilize targeted exome sequencing in Indian RB patients affected with unilateral non-familial RB. 75 unilateral RB patients below 5 years of age were enrolled. Genomic DNA was extracted from blood and tumor tissue. From peripheral blood DNA, all coding and exon/intron regions were amplified using PCR and direct sequencing. Cases which did not harbor pathogenic variants in peripheral blood DNA were further screened for mutations in their tumor tissue DNA using targeted exome sequencing. Three pathogenicity prediction tools (Mutation Taster, SIFT, and PolyPhen-2) were used to determine the pathogenicity of non-synonymous variations. An in-house bioinformatics pipeline was devised for the mutation screening by targeted exome sequencing. Protein modeling studies were also done to predict the effect of the mutations on the protein structure and function. RESULTS Using the mentioned approach, we found two novel variants (g.69673_69674insT and g.48373314C>A) in RB1 gene in peripheral blood DNA. We also found novel variants in eight genes (RB1, ACAD11, GPR151, KCNA1, OTOR, SOX30, ARL11, and MYCT1) that may be associated with RB pathogenesis. CONCLUSION The present study expands our current knowledge regarding the genomic landscape of RB and also highlights the importance of NGS technologies to detect genes and novel variants that may play an important role in cancer initiation, progression, and prognosis.
Collapse
Affiliation(s)
- Shilpa Bisht
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Bhavna Chawla
- Ocular Oncology Service, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Kumar
- Computational Genomics Centre, Indian Council of Medical Research, New Delhi, India
| | - Viswanathan Vijayan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Rima Dada
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
5
|
ARL11 correlates with the immunosuppression and poor prognosis in breast cancer: A comprehensive bioinformatics analysis of ARL family members. PLoS One 2022; 17:e0274757. [DOI: 10.1371/journal.pone.0274757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
ADP-ribosylation factor-like protein (ARL) family members (ARLs) may regulate the malignant phenotypes of cancer cells. However, relevant studies on ARLs in breast cancer (BC) are limited. In this research, the expression profiles, genetic variations, and prognostic values of ARLs in BC have been systematically analyzed for the first time using various databases. We find that ARLs are significantly dysregulated in BC according to the TCGA database, which may result from DNA methylation and copy number alteration. Prognostic analysis suggests that ARL11 is the most significant prognostic indicator for BC, and higher ARL11 predicts worse clinical outcomes for BC patients. Further functional enrichment analysis demonstrates that ARL11 enhances the immunosuppression in BC, and dysregulation of ARL11 is significantly associated with immune infiltration in various types of cancer. Our results demonstrate the potential of ARL11 as an immune therapeutic target for BC.
Collapse
|
6
|
Brito C, Costa-Silva B, Barral DC, Pojo M. Unraveling the Relevance of ARL GTPases in Cutaneous Melanoma Prognosis through Integrated Bioinformatics Analysis. Int J Mol Sci 2021; 22:9260. [PMID: 34502169 PMCID: PMC8431576 DOI: 10.3390/ijms22179260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
Cutaneous melanoma (CM) is the deadliest skin cancer, whose molecular pathways underlying its malignancy remain unclear. Therefore, new information to guide evidence-based clinical decisions is required. Adenosine diphosphate (ADP)-ribosylation factor-like (ARL) proteins are membrane trafficking regulators whose biological relevance in CM is undetermined. Here, we investigated ARL expression and its impact on CM prognosis and immune microenvironment through integrated bioinformatics analysis. Our study found that all 22 ARLs are differentially expressed in CM. Specifically, ARL1 and ARL11 are upregulated and ARL15 is downregulated regardless of mutational frequency or copy number variations. According to TCGA data, ARL1 and ARL15 represent independent prognostic factors in CM as well as ARL11 based on GEPIA and OncoLnc. To investigate the mechanisms by which ARL1 and ARL11 increase patient survival while ARL15 reduces it, we evaluated their correlation with the immune microenvironment. CD4+ T cells and neutrophil infiltrates are significantly increased by ARL1 expression. Furthermore, ARL11 expression was correlated with 17 out of 21 immune infiltrates, including CD8+ T cells and M2 macrophages, described as having anti-tumoral activity. Likewise, ARL11 is interconnected with ZAP70, ADAM17, and P2RX7, which are implicated in immune cell activation. Collectively, this study provides the first evidence that ARL1, ARL11, and ARL15 may influence CM progression, prognosis, and immune microenvironment remodeling.
Collapse
Affiliation(s)
- Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM) do Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal;
| | - Bruno Costa-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Avenida de Brasília, 1400-038 Lisbon, Portugal;
| | - Duarte C. Barral
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal;
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM) do Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal;
| |
Collapse
|
7
|
Casalou C, Ferreira A, Barral DC. The Role of ARF Family Proteins and Their Regulators and Effectors in Cancer Progression: A Therapeutic Perspective. Front Cell Dev Biol 2020; 8:217. [PMID: 32426352 PMCID: PMC7212444 DOI: 10.3389/fcell.2020.00217] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
The Adenosine diphosphate-Ribosylation Factor (ARF) family belongs to the RAS superfamily of small GTPases and is involved in a wide variety of physiological processes, such as cell proliferation, motility and differentiation by regulating membrane traffic and associating with the cytoskeleton. Like other members of the RAS superfamily, ARF family proteins are activated by Guanine nucleotide Exchange Factors (GEFs) and inactivated by GTPase-Activating Proteins (GAPs). When active, they bind effectors, which mediate downstream functions. Several studies have reported that cancer cells are able to subvert membrane traffic regulators to enhance migration and invasion. Indeed, members of the ARF family, including ARF-Like (ARL) proteins have been implicated in tumorigenesis and progression of several types of cancer. Here, we review the role of ARF family members, their GEFs/GAPs and effectors in tumorigenesis and cancer progression, highlighting the ones that can have a pro-oncogenic behavior or function as tumor suppressors. Moreover, we propose possible mechanisms and approaches to target these proteins, toward the development of novel therapeutic strategies to impair tumor progression.
Collapse
Affiliation(s)
- Cristina Casalou
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Andreia Ferreira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Duarte C Barral
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
8
|
Guo F, Yuan D, Zhang J, Zhang H, Wang C, Zhu L, Zhang J, Pan Y, Shao C. Silencing of ARL14 Gene Induces Lung Adenocarcinoma Cells to a Dormant State. Front Cell Dev Biol 2019; 7:238. [PMID: 31750299 PMCID: PMC6843082 DOI: 10.3389/fcell.2019.00238] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022] Open
Abstract
Recently, a growing number of ADP ribosylation factor (ARF) family members has been suggested to be critical in tumorigenesis. However, the effects of most ARF members on lung adenocarcinoma pathogenesis are still not well disclosed yet. In this study, ARF-like GTPase 14 (ARL14) was screened as an important prognostic factor of lung adenocarcinoma from The Cancer Genome Atlas (TCGA) database and validated by our in vitro experiments. It was found that silencing of ARL14 gene inhibited cell proliferation and the abilities of cell migration and invasion, and it also attenuated radiation damage of lung adenocarcinoma cells but had no effect on the proliferation of normal lung cells. Notably, ARL14 siRNA blocked the extracellular signal-regulated kinase (ERK)/p38 signaling pathway and induced cell cycle arrest in G0 phase, ultimately leading to cell dormancy. Moreover, ARL14 siRNA enhanced the expression of cell death activator DFFA-like effector (CIDEC) that had opposite roles in cell proliferation and migration to ALR14. Collectively, our results suggest that ARL14 has an important role in the pathogenesis of lung adenocarcinoma through CIDEC/ERK/p38 signaling pathway, and thus it could be applied as a new candidate of prognosis indicator and/or therapeutic target of lung adenocarcinoma.
Collapse
Affiliation(s)
- Fei Guo
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Dexiao Yuan
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Junling Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Hang Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Chen Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Lin Zhu
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Yan Pan
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Sztul E, Chen PW, Casanova JE, Cherfils J, Dacks JB, Lambright DG, Lee FJS, Randazzo PA, Santy LC, Schürmann A, Wilhelmi I, Yohe ME, Kahn RA. ARF GTPases and their GEFs and GAPs: concepts and challenges. Mol Biol Cell 2019; 30:1249-1271. [PMID: 31084567 PMCID: PMC6724607 DOI: 10.1091/mbc.e18-12-0820] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/26/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Detailed structural, biochemical, cell biological, and genetic studies of any gene/protein are required to develop models of its actions in cells. Studying a protein family in the aggregate yields additional information, as one can include analyses of their coevolution, acquisition or loss of functionalities, structural pliability, and the emergence of shared or variations in molecular mechanisms. An even richer understanding of cell biology can be achieved through evaluating functionally linked protein families. In this review, we summarize current knowledge of three protein families: the ARF GTPases, the guanine nucleotide exchange factors (ARF GEFs) that activate them, and the GTPase-activating proteins (ARF GAPs) that have the ability to both propagate and terminate signaling. However, despite decades of scrutiny, our understanding of how these essential proteins function in cells remains fragmentary. We believe that the inherent complexity of ARF signaling and its regulation by GEFs and GAPs will require the concerted effort of many laboratories working together, ideally within a consortium to optimally pool information and resources. The collaborative study of these three functionally connected families (≥70 mammalian genes) will yield transformative insights into regulation of cell signaling.
Collapse
Affiliation(s)
- Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Pei-Wen Chen
- Department of Biology, Williams College, Williamstown, MA 01267
| | - James E. Casanova
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - Jacqueline Cherfils
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS and Ecole Normale Supérieure Paris-Saclay, 94235 Cachan, France
| | - Joel B. Dacks
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - David G. Lambright
- Program in Molecular Medicine and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Amherst, MA 01605
| | - Fang-Jen S. Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | | | - Lorraine C. Santy
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Annette Schürmann
- German Institute of Human Nutrition, 85764 Potsdam-Rehbrücke, Germany
| | - Ilka Wilhelmi
- German Institute of Human Nutrition, 85764 Potsdam-Rehbrücke, Germany
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322-3050
| |
Collapse
|
10
|
Detection of novel fusion-transcripts by RNA-Seq in T-cell lymphoblastic lymphoma. Sci Rep 2019; 9:5179. [PMID: 30914738 PMCID: PMC6435891 DOI: 10.1038/s41598-019-41675-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Fusions transcripts have been proven to be strong drivers for neoplasia-associated mutations, although their incidence in T-cell lymphoblastic lymphoma needs to be determined yet. Using RNA-Seq we have selected 55 fusion transcripts identified by at least two of three detection methods in the same tumour. We confirmed the existence of 24 predicted novel fusions that had not been described in cancer or normal tissues yet, indicating the accuracy of the prediction. Of note, one of them involves the proto oncogene TAL1. Other confirmed fusions could explain the overexpression of driver genes such as COMMD3-BMI1, LMO1 or JAK3. Five fusions found exclusively in tumour samples could be considered pathogenic (NFYG-TAL1, RIC3-TCRBC2, SLC35A3-HIAT1, PICALM MLLT10 and MLLT10-PICALM). However, other fusions detected simultaneously in normal and tumour samples (JAK3-INSL3, KANSL1-ARL17A/B and TFG-ADGRG7) could be germ-line fusions genes involved in tumour-maintaining tasks. Notably, some fusions were confirmed in more tumour samples than predicted, indicating that the detection methods underestimated the real number of existing fusions. Our results highlight the potential of RNA-Seq to identify new cryptic fusions, which could be drivers or tumour-maintaining passenger genes. Such novel findings shed light on the searching for new T-LBL biomarkers in these haematological disorders.
Collapse
|
11
|
Platko K, Lebeau P, Austin RC. MAPping the kinase landscape of macrophage activation. J Biol Chem 2019; 293:9910-9911. [PMID: 29934367 DOI: 10.1074/jbc.h118.003380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ARL11 is a tumor suppressor gene with established pro-apoptotic properties, but its function beyond this role is poorly understood. A new analysis of macrophage activation has identified ARL11 as a novel regulator of a mitogen-activated protein kinase (MAPK). These findings expand on the function of ARL11 beyond its tumor suppressor activity and highlight a novel role as a regulator of macrophage activation and inflammatory response.
Collapse
Affiliation(s)
- Khrystyna Platko
- From the Department of Medicine, Division of Nephrology, McMaster University, St. Joseph's Healthcare Hamilton and Hamilton Center for Kidney Research, Hamilton, Ontario L8N 4A6 and
| | - Paul Lebeau
- From the Department of Medicine, Division of Nephrology, McMaster University, St. Joseph's Healthcare Hamilton and Hamilton Center for Kidney Research, Hamilton, Ontario L8N 4A6 and
| | - Richard C Austin
- From the Department of Medicine, Division of Nephrology, McMaster University, St. Joseph's Healthcare Hamilton and Hamilton Center for Kidney Research, Hamilton, Ontario L8N 4A6 and .,Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton Health Sciences and McMaster University, Hamilton, Ontario L8L 2X2, Canada
| |
Collapse
|
12
|
Arya SB, Kumar G, Kaur H, Kaur A, Tuli A. ARL11 regulates lipopolysaccharide-stimulated macrophage activation by promoting mitogen-activated protein kinase (MAPK) signaling. J Biol Chem 2018; 293:9892-9909. [PMID: 29618517 PMCID: PMC6016484 DOI: 10.1074/jbc.ra117.000727] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/20/2018] [Indexed: 12/13/2022] Open
Abstract
ADP-ribosylation factor-like GTPase 11 (ARL11) is a cancer-predisposing gene that has remained functionally uncharacterized to date. In this study, we report that ARL11 is endogenously expressed in mouse and human macrophages and regulates their activation in response to lipopolysaccharide (LPS) stimulation. Accordingly, depletion of ARL11 impaired both LPS-stimulated pro-inflammatory cytokine production by macrophages and their ability to control intracellular replication of Salmonella. LPS-stimulated activation of extracellular signal–regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) was substantially compromised in Arl11-silenced macrophages. In contrast, increased expression of ARL11 led to constitutive ERK1/2 phosphorylation, resulting in macrophage exhaustion. Finally, we found that ARL11 forms a complex with phospho-ERK in macrophages within minutes of LPS stimulation. Taken together, our findings establish ARL11 as a novel regulator of ERK signaling in macrophages, required for macrophage activation and immune function.
Collapse
Affiliation(s)
- Subhash B Arya
- From the Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Gaurav Kumar
- From the Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Harmeet Kaur
- From the Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Amandeep Kaur
- From the Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Amit Tuli
- From the Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| |
Collapse
|
13
|
Jiang Y, Zhao CY, Cheng LC, Xu B, Lv HY. ARLTS1 polymorphism is associated with an increased risk of familial cancer: evidence from a meta-analysis. Hered Cancer Clin Pract 2017. [PMID: 28630657 PMCID: PMC5470195 DOI: 10.1186/s13053-017-0068-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Adenosine diphosphate (ADP)-ribosylation factor-like tumour suppressor gene 1(ARLTS1) might be associated with an increased risk of several types of familial cancers. However, previous studies have shown that cancer susceptibility is not completely consistent with ARLTS1 polymorphisms, and the precise mechanism remains unknown. Therefore, we conducted a meta-analysis of case-control studies by searching the PubMed, Embase, OVID, Science Direct and Chinese National Knowledge Infrastructure (CNKI) databases. In total, 12 studies met the inclusion criteria and were included in this meta-analysis. Statistical analyses were performed using STATA 11.0 software. Overall, the Cys148Arg T > C variant significantly increased cancer risk (CC vs. TT: OR = 1.27, 95% CI = 1.15–1.41, P < 0.05). The stratification indicated that the Cys148Arg variant is significantly associated with sporadic cancer (CC vs. TT: OR = 1.36, 95% CI = 1.18–1.55) and familial cancer (CC vs. TT: OR = 1.26, 95% CI = 1.12–1.43). Trp149Stop, Pro131Leu, Ser99Ser and Leu132Leu were not correlated with cancer susceptibility. Based on these results, we demonstrated that the ARLTS1 Cys148Arg polymorphism is associated with an increased risk of sporadic cancer and familial cancer, and there were no associations between the other four SNPs (i.e., Trp149Stop, Pro131Leu, Ser99Ser and Leu132Leu) and cancer risk.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Pharmaceuticals, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 People's Republic of China
| | - Chen-Yang Zhao
- Department of Pharmaceuticals, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 People's Republic of China
| | - Li-Chun Cheng
- Department of Pharmaceuticals, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 People's Republic of China
| | - Bing Xu
- Department of Pharmaceuticals, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 People's Republic of China
| | - Hui-Yi Lv
- Department of Pharmaceuticals, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 People's Republic of China
| |
Collapse
|
14
|
Zhou JX, Yang X, Ning S, Wang L, Wang K, Zhang Y, Yuan F, Li F, Zhuo DD, Tang L, Zhuo D. Identification of KANSARL as the first cancer predisposition fusion gene specific to the population of European ancestry origin. Oncotarget 2017; 8:50594-50607. [PMID: 28881586 PMCID: PMC5584173 DOI: 10.18632/oncotarget.16385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Gene fusion is one of the hallmarks of cancer. Recent advances in RNA-seq of cancer transcriptomes have facilitated the discovery of fusion transcripts. In this study, we report identification of a surprisingly large number of fusion transcripts, including six KANSARL (KANSL1-ARL17A) transcripts that resulted from the fusion between the KANSL1 and ARL17A genes using a RNA splicingcode model. Five of these six KANSARL fusion transcripts are novel. By systematic analysis of RNA-seq data of glioblastoma, prostate cancer, lung cancer, breast cancer, and lymphoma from different regions of the World, we have found that KANSARL fusion transcripts were rarely detected in the tumors of individuals from Asia or Africa. In contrast, they exist in 30 - 52% of the tumors from North Americans cancer patients. Analysis of CEPH/Utah Pedigree 1463 has revealed that KANSARL is a familially-inherited fusion gene. Further analysis of RNA-seq datasets of the 1000 Genome Project has indicated that KANSARL fusion gene is specific to 28.9% of the population of European ancestry origin. In summary, we demonstrated that KANSARL is the first cancer predisposition fusion gene associated with genetic backgrounds of European ancestry origin.
Collapse
Affiliation(s)
- Jeff Xiwu Zhou
- Department of Medicine, School of Medicine, Ningbo University, Ningbo, China
| | - Xiaoyan Yang
- SplicingCodes.com, Biotailor Inc., Palmetto Bay, FL, USA
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, East Tennessee State University, Johnson City, TN, USA
| | - Yanbin Zhang
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| | - Fenghua Yuan
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| | - Fengli Li
- Department of Medicine, School of Medicine, Ningbo University, Ningbo, China
| | - David D Zhuo
- SplicingCodes.com, Biotailor Inc., Palmetto Bay, FL, USA
| | - Liren Tang
- SplicingCodes.com, Biotailor Inc., Palmetto Bay, FL, USA
| | - Degen Zhuo
- SplicingCodes.com, Biotailor Inc., Palmetto Bay, FL, USA
| |
Collapse
|
15
|
Hamadou WS, Besbes S, Mani R, Bourdon V, Ben Youssef Y, Achour B, Regaieg H, Eisinger F, Mari V, Gesta P, Dreyfus H, Bonadona V, Dugast C, Zattara H, Faivre L, Noguchi T, Khélif A, Sobol H, Soua Z. ARLTS1, potential candidate gene in familial aggregation of hematological malignancies. Bull Cancer 2016; 104:123-127. [PMID: 27866680 DOI: 10.1016/j.bulcan.2016.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Genetic predisposition to familial hematological malignancies was previously described through several epidemiological analyses, but the genetic basis remains unclear. The tumor-suppressor ARLTS1 gene was previously described in sporadic hematological malignancies and familial cancer context. METHODS In this study, we sequence the ARLTS1 gene in 100 patients belonging to 88 independent Tunisian and French families. RESULTS After gene sequencing, we report 8 genetic variations, most of which were previously reported in several cancer forms. The most common variants were W149X and C148R and were previously associated to B-cell chronic lymphocytic leukemia and to high-risk of familial breast cancer. CONCLUSIONS These results emphasize the fact that ARLTS1 gene mutations can be considered as a potential predisposing factor in familial hematological malignancies and other several cancer forms.
Collapse
Affiliation(s)
- Walid Sabri Hamadou
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia.
| | - Sawsen Besbes
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia
| | - Rahma Mani
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia
| | - Violaine Bourdon
- Institut Paoli-Calmettes, département d'oncologie génétique, de prévention et dépistage, 232, boulevard de Sainte-Marguerite, 13009 Marseille, France
| | - Yosra Ben Youssef
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia; Service d'hématologie clinique, CHU Farhat Hached, Sousse, Tunisia
| | - Béchir Achour
- Service d'hématologie clinique, CHU Farhat Hached, Sousse, Tunisia
| | - Haifa Regaieg
- Service d'hématologie clinique, CHU Farhat Hached, Sousse, Tunisia
| | - François Eisinger
- Institut Paoli-Calmettes, centre de lutte contre le cancer, département d'anticipation et de suivi du cancer, 232, boulevard de Sainte-Marguerite, 13009 Marseille, France
| | - Véronique Mari
- Centre de lutte contre le cancer, centre Antoine-Lacassagne, service d'oncologie génétique, 33, avenue de Valombrose, 06100 Nice, France
| | - Paul Gesta
- Centre hospitalier Georges-Renon, service oncogénétique pour la consultation oncogénétique régionale Poitou-Charentes, 79021 Niort, France
| | - Hélène Dreyfus
- Institut Sainte-Catherine, 250, chemin de Baigne-Pieds, 84918 Avignon cedex 9, France
| | - Valérie Bonadona
- Centre Léon-Bérard, unité de génétique épidémiologique, 28, prom. Léa-et-Napoléon-Bullukian, 69008 Lyon, France
| | - Catherine Dugast
- Centre Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France
| | - Hélène Zattara
- Hôpital de la Timone, département de génétique, 264, rue Saint-Pierre, 13385 Marseille, France
| | - Laurence Faivre
- CHU de Dijon, hôpital d'Enfants, 14, rue Paul-Gaffarel, 21079 Dijon, France
| | - Testsuro Noguchi
- Institut Paoli-Calmettes, département d'oncologie génétique, de prévention et dépistage, 232, boulevard de Sainte-Marguerite, 13009 Marseille, France
| | - Abderrahim Khélif
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia; Service d'hématologie clinique, CHU Farhat Hached, Sousse, Tunisia
| | - Hagay Sobol
- Institut Paoli-Calmettes, département d'oncologie génétique, de prévention et dépistage, 232, boulevard de Sainte-Marguerite, 13009 Marseille, France
| | - Zohra Soua
- Université de Sousse, faculté de médecine de Sousse, laboratoire de Biochimie, UR « biologie moléculaire des leucémies et lymphomes », avenue Mohamed Karoui, 4000 Sousse, Tunisia
| |
Collapse
|
16
|
Higgins MJ, Serrano A, Boateng KY, Parsons VA, Phuong T, Seifert A, Ricca JM, Tucker KC, Eidelman AS, Carey MA, Kurt RA. A Multifaceted Role for Myd88-Dependent Signaling in Progression of Murine Mammary Carcinoma. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:157-167. [PMID: 27812285 PMCID: PMC5084708 DOI: 10.4137/bcbcr.s40075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/10/2016] [Accepted: 07/12/2016] [Indexed: 02/06/2023]
Abstract
Previous data obtained in our laboratory suggested that there may be constitutive signaling through the myeloid differentiation primary response gene 88 (Myd88)-dependent signaling cascade in murine mammary carcinoma. Here, we extended these findings by showing that, in the absence of an added Toll-like receptor (TLR) agonist, the myddosome complex was preformed in 4T1 tumor cells, and that Myd88 influenced cytoplasmic extracellular signal–regulated kinase (Erk)1/Erk2 levels, nuclear levels of nuclear factor-kappaB (NFκB) and signal transducer and activator of transcription 5 (STAT5), tumor-derived chemokine (C–C motif) ligand 2 (CCL2) expression, and in vitro and in vivo tumor growth. In addition, RNA-sequencing revealed that Myd88-dependent signaling enhanced the expression of genes that could contribute to breast cancer progression and genes previously associated with poor outcome for patients with breast cancer, in addition to suppressing the expression of genes capable of inhibiting breast cancer progression. Yet, Myd88-dependent signaling in tumor cells also suppressed expression of genes that could contribute to tumor progression. Collectively, these data revealed a multifaceted role for Myd88-dependent signaling in murine mammary carcinoma.
Collapse
Affiliation(s)
- Mary J Higgins
- Department of Biology, Lafayette College, Easton, PA, USA
| | | | - Kofi Y Boateng
- Department of Biology, Lafayette College, Easton, PA, USA
| | | | - Tiffany Phuong
- Department of Biology, Lafayette College, Easton, PA, USA
| | - Alyssa Seifert
- Department of Biology, Lafayette College, Easton, PA, USA
| | - Jacob M Ricca
- Department of Biology, Lafayette College, Easton, PA, USA
| | - Kyle C Tucker
- Department of Biology, Lafayette College, Easton, PA, USA
| | | | | | - Robert A Kurt
- Department of Biology, Lafayette College, Easton, PA, USA
| |
Collapse
|
17
|
Thériault BL, Dimaras H, Gallie BL, Corson TW. The genomic landscape of retinoblastoma: a review. Clin Exp Ophthalmol 2013; 42:33-52. [PMID: 24433356 DOI: 10.1111/ceo.12132] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/07/2013] [Indexed: 12/13/2022]
Abstract
Retinoblastoma is a paediatric ocular tumour that continues to reveal much about the genetic basis of cancer development. Study of genomic aberrations in retinoblastoma tumours has exposed important mechanisms of cancer development and identified oncogenes and tumour suppressors that offer potential points of therapeutic intervention. The recent development of next-generation genomic technologies has allowed further refinement of the genomic landscape of retinoblastoma at high resolution. In a relatively short period of time, a wealth of genetic and epigenetic data has emerged on a small number of tumour samples. These data highlight the inherent molecular complexity of this cancer despite the fact that most retinoblastomas are initiated by the inactivation of a single tumour suppressor gene. This review outlines the current understanding of the genomic, genetic and epigenetic changes in retinoblastoma, highlighting recent genome-wide analyses that have identified exciting candidate genes worthy of further validation as potential prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Brigitte L Thériault
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
18
|
Mercer SE, Cheng CH, Atkinson DL, Krcmery J, Guzman CE, Kent DT, Zukor K, Marx KA, Odelberg SJ, Simon HG. Multi-tissue microarray analysis identifies a molecular signature of regeneration. PLoS One 2012; 7:e52375. [PMID: 23300656 PMCID: PMC3530543 DOI: 10.1371/journal.pone.0052375] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 11/14/2012] [Indexed: 02/06/2023] Open
Abstract
The inability to functionally repair tissues that are lost as a consequence of disease or injury remains a significant challenge for regenerative medicine. The molecular and cellular processes involved in complete restoration of tissue architecture and function are expected to be complex and remain largely unknown. Unlike humans, certain salamanders can completely regenerate injured tissues and lost appendages without scar formation. A parsimonious hypothesis would predict that all of these regenerative activities are regulated, at least in part, by a common set of genes. To test this hypothesis and identify genes that might control conserved regenerative processes, we performed a comprehensive microarray analysis of the early regenerative response in five regeneration-competent tissues from the newt Notophthalmus viridescens. Consistent with this hypothesis, we established a molecular signature for regeneration that consists of common genes or gene family members that exhibit dynamic differential regulation during regeneration in multiple tissue types. These genes include members of the matrix metalloproteinase family and its regulators, extracellular matrix components, genes involved in controlling cytoskeleton dynamics, and a variety of immune response factors. Gene Ontology term enrichment analysis validated and supported their functional activities in conserved regenerative processes. Surprisingly, dendrogram clustering and RadViz classification also revealed that each regenerative tissue had its own unique temporal expression profile, pointing to an inherent tissue-specific regenerative gene program. These new findings demand a reconsideration of how we conceptualize regenerative processes and how we devise new strategies for regenerative medicine.
Collapse
Affiliation(s)
- Sarah E. Mercer
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - Chia-Ho Cheng
- Department of Chemistry, University of Massachusetts-Lowell, Lowell, Massachusetts, United States of America
| | - Donald L. Atkinson
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Jennifer Krcmery
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - Claudia E. Guzman
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - David T. Kent
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Katherine Zukor
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Kenneth A. Marx
- Department of Chemistry, University of Massachusetts-Lowell, Lowell, Massachusetts, United States of America
| | - Shannon J. Odelberg
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hans-Georg Simon
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| |
Collapse
|
19
|
Abulí A, Fernández-Rozadilla C, Alonso-Espinaco V, Muñoz J, Gonzalo V, Bessa X, González D, Clofent J, Cubiella J, Morillas JD, Rigau J, Latorre M, Fernández-Bañares F, Peña E, Riestra S, Payá A, Jover R, Xicola RM, Llor X, Carvajal-Carmona L, Villanueva CM, Moreno V, Piqué JM, Carracedo A, Castells A, Andreu M, Ruiz-Ponte C, Castellví-Bel S. Case-control study for colorectal cancer genetic susceptibility in EPICOLON: previously identified variants and mucins. BMC Cancer 2011; 11:339. [PMID: 21819567 PMCID: PMC3176240 DOI: 10.1186/1471-2407-11-339] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 08/05/2011] [Indexed: 11/10/2022] Open
Abstract
Background Colorectal cancer (CRC) is the second leading cause of cancer death in developed countries. Familial aggregation in CRC is also important outside syndromic forms and, in this case, a polygenic model with several common low-penetrance alleles contributing to CRC genetic predisposition could be hypothesized. Mucins and GALNTs (N-acetylgalactosaminyltransferase) are interesting candidates for CRC genetic susceptibility and have not been previously evaluated. We present results for ten genetic variants linked to CRC risk in previous studies (previously identified category) and 18 selected variants from the mucin gene family in a case-control association study from the Spanish EPICOLON consortium. Methods CRC cases and matched controls were from EPICOLON, a prospective, multicenter, nationwide Spanish initiative, comprised of two independent stages. Stage 1 corresponded to 515 CRC cases and 515 controls, whereas stage 2 consisted of 901 CRC cases and 909 controls. Also, an independent cohort of 549 CRC cases and 599 controls outside EPICOLON was available for additional replication. Genotyping was performed for ten previously identified SNPs in ADH1C, APC, CCDN1, IL6, IL8, IRS1, MTHFR, PPARG, VDR and ARL11, and 18 selected variants in the mucin gene family. Results None of the 28 SNPs analyzed in our study was found to be associated with CRC risk. Although four SNPs were significant with a P-value < 0.05 in EPICOLON stage 1 [rs698 in ADH1C (OR = 1.63, 95% CI = 1.06-2.50, P-value = 0.02, recessive), rs1800795 in IL6 (OR = 1.62, 95% CI = 1.10-2.37, P-value = 0.01, recessive), rs3803185 in ARL11 (OR = 1.58, 95% CI = 1.17-2.15, P-value = 0.007, codominant), and rs2102302 in GALNTL2 (OR = 1.20, 95% CI = 1.00-1.44, P-value = 0.04, log-additive 0, 1, 2 alleles], only rs3803185 achieved statistical significance in EPICOLON stage 2 (OR = 1.34, 95% CI = 1.06-1.69, P-value = 0.01, recessive). In the joint analysis for both stages, results were only significant for rs3803185 (OR = 1.12, 95% CI = 1.00-1.25, P-value = 0.04, log-additive 0, 1, 2 alleles) and borderline significant for rs698 and rs2102302. The rs3803185 variant was not significantly associated with CRC risk in an external cohort (MCC-Spain), but it still showed some borderline significance in the pooled analysis of both cohorts (OR = 1.08, 95% CI = 0.98-1.18, P-value = 0.09, log-additive 0, 1, 2 alleles). Conclusions ARL11, ADH1C, GALNTL2 and IL6 genetic variants may have an effect on CRC risk. Further validation and meta-analyses should be undertaken in larger CRC studies.
Collapse
Affiliation(s)
- Anna Abulí
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, University of Barcelona, Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhou X, Münger K. Clld7, a candidate tumor suppressor on chromosome 13q14, regulates pathways of DNA damage/repair and apoptosis. Cancer Res 2010; 70:9434-43. [PMID: 20926398 DOI: 10.1158/0008-5472.can-10-1960] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic lymphocytic leukemia deletion gene 7 (Clld7) is a candidate tumor suppressor on chromosome 13q14. Clld7 encodes an evolutionarily conserved protein that contains an RCC1 domain plus broad complex, tramtrack, bric-a-brac (BTB), and POZ domains. In this study, we investigated the biological functions of Clld7 protein in inducible osteosarcoma cell lines. Clld7 induction inhibited cell growth, decreased cell viability, and increased γ-H2AX staining under conditions of caspase inhibition, indicating activation of the DNA damage/repair pathway. Real-time PCR analysis in tumor cells and normal human epithelial cells revealed Clld7 target genes that regulate DNA repair responses. Furthermore, depletion of Clld7 in normal human epithelial cells conferred resistance to apoptosis triggered by DNA damage. Taken together, the biological actions of Clld7 are consistent with those of a tumor suppressor.
Collapse
Affiliation(s)
- Xiaobo Zhou
- The Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
21
|
Sampieri K, Amenduni M, Papa FT, Katzaki E, Mencarelli MA, Marozza A, Epistolato MC, Toti P, Lazzi S, Bruttini M, De Filippis R, De Francesco S, Longo I, Meloni I, Mari F, Acquaviva A, Hadjistilianou T, Renieri A, Ariani F. Array comparative genomic hybridization in retinoma and retinoblastoma tissues. Cancer Sci 2009; 100:465-71. [PMID: 19183342 PMCID: PMC11159683 DOI: 10.1111/j.1349-7006.2008.01070.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In retinoblastoma, two RB1 mutations are necessary for tumor development. Recurrent genomic rearrangements may represent subsequent events required for retinoblastoma progression. Array-comparative genomic hybridization was carried out in 18 eye samples, 10 from bilateral and eight from unilateral retinoblastoma patients. Two unilateral cases also showed areas of retinoma. The most frequent imbalance in retinoblastomas was 6p gain (40%), followed by gains at 1q12-q25.3, 2p24.3-p24.2, 9q22.2, and 9q33.1 and losses at 11q24.3, 13q13.2-q22.3, and 16q12.1-q21. Bilateral cases showed a lower number of imbalances than unilateral cases (P = 0.002). Unilateral cases were divided into low-level (< or = 4) and high-level (> or = 7) chromosomal instability groups. The first group presented with younger age at diagnosis (mean 511 days) compared with the second group (mean 1606 days). In one retinoma case ophthalmoscopically diagnosed as a benign lesion no rearrangements were detected, whereas the adjacent retinoblastoma displayed seven aberrations. The other retinoma case identified by retrospective histopathological examination shared three rearrangements with the adjacent retinoblastoma. Two other gene-free rearrangements were retinoma specific. One rearrangement, dup5p, was retinoblastoma specific and included the SKP2 gene. Genomic profiling indicated that the first retinoma was a pretumoral lesion, whereas the other represents a subclone of cells bearing 'benign' rearrangements overwhelmed by another subclone presenting aberrations with higher 'oncogenic' potential. In summary, the present study shows that bilateral and unilateral retinoblastoma have different chromosomal instability that correlates with the age of tumor onset in unilateral cases. This is the first report of genomic profiling in retinoma tissue, shedding light on the different nature of lesions named 'retinoma'.
Collapse
Affiliation(s)
- Katia Sampieri
- Medical Genetics, Department of Molecular Biology, University of Siena, Policlinico Le Scotte, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|