51
|
Stojic L, Niemczyk M, Orjalo A, Ito Y, Ruijter AEM, Uribe-Lewis S, Joseph N, Weston S, Menon S, Odom DT, Rinn J, Gergely F, Murrell A. Transcriptional silencing of long noncoding RNA GNG12-AS1 uncouples its transcriptional and product-related functions. Nat Commun 2016; 7:10406. [PMID: 26832224 PMCID: PMC4740813 DOI: 10.1038/ncomms10406] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/08/2015] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) regulate gene expression via their RNA product or through transcriptional interference, yet a strategy to differentiate these two processes is lacking. To address this, we used multiple small interfering RNAs (siRNAs) to silence GNG12-AS1, a nuclear lncRNA transcribed in an antisense orientation to the tumour-suppressor DIRAS3. Here we show that while most siRNAs silence GNG12-AS1 post-transcriptionally, siRNA complementary to exon 1 of GNG12-AS1 suppresses its transcription by recruiting Argonaute 2 and inhibiting RNA polymerase II binding. Transcriptional, but not post-transcriptional, silencing of GNG12-AS1 causes concomitant upregulation of DIRAS3, indicating a function in transcriptional interference. This change in DIRAS3 expression is sufficient to impair cell cycle progression. In addition, the reduction in GNG12-AS1 transcripts alters MET signalling and cell migration, but these are independent of DIRAS3. Thus, differential siRNA targeting of a lncRNA allows dissection of the functions related to the process and products of its transcription.
Collapse
Affiliation(s)
- Lovorka Stojic
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Malwina Niemczyk
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Arturo Orjalo
- Biosearch Technologies Inc., 2199S. McDowell Boulevard, Petaluma, California 94954, USA
| | - Yoko Ito
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Anna Elisabeth Maria Ruijter
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Santiago Uribe-Lewis
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Nimesh Joseph
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Stephen Weston
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Suraj Menon
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Duncan T. Odom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - John Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Adele Murrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| |
Collapse
|
52
|
Kok DEG, Dhonukshe-Rutten RAM, Lute C, Heil SG, Uitterlinden AG, van der Velde N, van Meurs JBJ, van Schoor NM, Hooiveld GJEJ, de Groot LCPGM, Kampman E, Steegenga WT. The effects of long-term daily folic acid and vitamin B12 supplementation on genome-wide DNA methylation in elderly subjects. Clin Epigenetics 2015; 7:121. [PMID: 26568774 PMCID: PMC4644301 DOI: 10.1186/s13148-015-0154-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/04/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Folate and its synthetic form folic acid function as donor of one-carbon units and have been, together with other B-vitamins, implicated in programming of epigenetic processes such as DNA methylation during early development. To what extent regulation of DNA methylation can be altered via B-vitamins later in life, and how this relates to health and disease, is not exactly known. The aim of this study was to identify effects of long-term supplementation with folic acid and vitamin B12 on genome-wide DNA methylation in elderly subjects. This project was part of a randomized, placebo-controlled trial on effects of supplemental intake of folic acid and vitamin B12 on bone fracture incidence (B-vitamins for the PRevention Of Osteoporotic Fractures (B-PROOF) study). Participants with mildly elevated homocysteine levels, aged 65-75 years, were randomly assigned to take 400 μg folic acid and 500 μg vitamin B12 per day or a placebo during an intervention period of 2 years. DNA was isolated from buffy coats, collected before and after intervention, and genome-wide DNA methylation was determined in 87 participants (n = 44 folic acid/vitamin B12, n = 43 placebo) using the Infinium HumanMethylation450 BeadChip. RESULTS After intervention with folic acid and vitamin B12, 162 (versus 14 in the placebo group) of the 431,312 positions were differentially methylated as compared to baseline. Comparisons of the DNA methylation changes in the participants receiving folic acid and vitamin B12 versus placebo revealed one single differentially methylated position (cg19380919) with a borderline statistical significance. However, based on the analyses of differentially methylated regions (DMRs) consisting of multiple positions, we identified 6 regions that differed statistically significantly between the intervention and placebo group. Pronounced changes were found for regions in the DIRAS3, ARMC8, and NODAL genes, implicated in carcinogenesis and early embryonic development. Furthermore, serum levels of folate and vitamin B12 or plasma homocysteine were related to DNA methylation of 173, 425, and 11 regions, respectively. Interestingly, for several members of the developmental HOX genes, DNA methylation was related to serum levels of folate. CONCLUSIONS Long-term supplementation with folic acid and vitamin B12 in elderly subjects resulted in effects on DNA methylation of several genes, among which genes implicated in developmental processes.
Collapse
Affiliation(s)
- Dieuwertje E G Kok
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | | | - Carolien Lute
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Sandra G Heil
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands ; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nathalie van der Velde
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands ; Department of Internal Medicine, Section of Geriatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - Joyce B J van Meurs
- Genetic Laboratory Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Guido J E J Hooiveld
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Lisette C P G M de Groot
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Ellen Kampman
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| | - Wilma T Steegenga
- Division of Human Nutrition, Wageningen University, PO Box 8129, 6700 EV Wageningen, The Netherlands
| |
Collapse
|
53
|
ARHI (DIRAS3)-mediated autophagy-associated cell death enhances chemosensitivity to cisplatin in ovarian cancer cell lines and xenografts. Cell Death Dis 2015; 6:e1836. [PMID: 26247722 PMCID: PMC4558501 DOI: 10.1038/cddis.2015.208] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 05/24/2015] [Accepted: 06/26/2015] [Indexed: 11/08/2022]
Abstract
Autophagy can sustain or kill tumor cells depending upon the context. The mechanism of autophagy-associated cell death has not been well elucidated and autophagy has enhanced or inhibited sensitivity of cancer cells to cytotoxic chemotherapy in different models. ARHI (DIRAS3), an imprinted tumor suppressor gene, is downregulated in 60% of ovarian cancers. In cell culture, re-expression of ARHI induces autophagy and ovarian cancer cell death within 72 h. In xenografts, re-expression of ARHI arrests cell growth and induces autophagy, but does not kill engrafted cancer cells. When ARHI levels are reduced after 6 weeks, dormancy is broken and xenografts grow promptly. In this study, ARHI-induced ovarian cancer cell death in culture has been found to depend upon autophagy and has been linked to G1 cell-cycle arrest, enhanced reactive oxygen species (ROS) activity, RIP1/RIP3 activation and necrosis. Re-expression of ARHI enhanced the cytotoxic effect of cisplatin in cell culture, increasing caspase-3 activation and PARP cleavage by inhibiting ERK and HER2 activity and downregulating XIAP and Bcl-2. In xenografts, treatment with cisplatin significantly slowed the outgrowth of dormant autophagic cells after reduction of ARHI, but the addition of chloroquine did not further inhibit xenograft outgrowth. Taken together, we have found that autophagy-associated cancer cell death and autophagy-enhanced sensitivity to cisplatin depend upon different mechanisms and that dormant, autophagic cancer cells are still vulnerable to cisplatin-based chemotherapy.
Collapse
|
54
|
Ye K, Wang S, Yang Y, Kang X, Wang J, Han H. Aplasia Ras homologue member Ⅰ overexpression inhibits tumor growth and induces apoptosis through inhibition of PI3K/Akt survival pathways in human osteosarcoma MG-63 cells in culture. Int J Mol Med 2015; 36:776-82. [PMID: 26165148 DOI: 10.3892/ijmm.2015.2278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/26/2015] [Indexed: 11/06/2022] Open
Abstract
Aplasia Ras homologue member Ⅰ (ARHI), an imprinted tumor-suppressor gene, is downregulated in various types of cancer. However, the expression, function and specific mechanisms of ARHI in human osteosarcoma (OS) cells remain unclear. The aim of the present study was to assess the effect of ARHI on OS cell proliferation and apoptosis and its associated mechanism. In the study, ARHI mRNA and protein levels were markedly downregulated in OS cells compared with the human osteoblast precursor cell line hFOB1.19. By generating stable transfectants, ARHI was overexpressed in OS cells that had low levels of ARHI. Overexpression of ARHI inhibited cell viability and proliferation and induced apoptosis. However, caspase‑3 activity was not changed by ARHI overexpression. In addition, phosphorylated Akt protein expression decreased in the ARHI overexpression group compared to that in the control vector group. The knockdown of ARHI also resulted in the promotion of cell proliferation and the attenuation of apoptosis in MG‑63 cells. Additionally, ARHI silencing increased the level of p‑Akt. The present results indicate that ARHI inhibits OS cell proliferation and may have a key role in the development of OS.
Collapse
Affiliation(s)
- Kaishan Ye
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Shuanke Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Yong Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xuewen Kang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Jing Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Hua Han
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
55
|
Kim CJ, Shin JW, Jung SW, Park BR, Park NH. Somatic Mutation of ARHI Gene in Hepatocellular Carcinomas. Pathol Oncol Res 2015; 21:1277-9. [PMID: 26143066 DOI: 10.1007/s12253-015-9924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 02/20/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Chang Jae Kim
- Biomedical Research Center, Ulsan University Hospital, College of Medicine, University of Ulsan, Ulsan, 682-714, Republic of Korea
| | | | | | | | | |
Collapse
|
56
|
Lu Z, Baquero MT, Yang H, Yang M, Reger AS, Kim C, Levine DA, Clarke CH, Liao WSL, Bast RC. DIRAS3 regulates the autophagosome initiation complex in dormant ovarian cancer cells. Autophagy 2015; 10:1071-92. [PMID: 24879154 DOI: 10.4161/auto.28577] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
DIRAS3 is an imprinted tumor suppressor gene that is downregulated in 60% of human ovarian cancers. Re-expression of DIRAS3 at physiological levels inhibits proliferation, decreases motility, induces autophagy, and regulates tumor dormancy. Functional inhibition of autophagy with choroquine in dormant xenografts that express DIRAS3 significantly delays tumor regrowth after DIRAS3 levels are reduced, suggesting that autophagy sustains dormant ovarian cancer cells. This study documents a newly discovered role for DIRAS3 in forming the autophagosome initiation complex (AIC) that contains BECN1, PIK3C3, PIK3R4, ATG14, and DIRAS3. Participation of BECN1 in the AIC is inhibited by binding of BECN1 homodimers to BCL2. DIRAS3 binds BECN1, disrupting BECN1 homodimers and displacing BCL2. Binding of DIRAS3 to BECN1 increases the association of BECN1 with PIK3C3 and ATG14, facilitating AIC activation. Amino acid starvation of cells induces DIRAS3 expression, reduces BECN1-BCL2 interaction and promotes autophagy, whereas DIRAS3 depletion blocks amino acid starvation-induced autophagy. In primary ovarian cancers, punctate expression of DIRAS3, BECN1, and the autophagic biomarker MAP1LC3 are highly correlated (P<0.0001), underlining the clinical relevance of these mechanistic studies. Punctate expression of DIRAS3 and MAP1LC3 was detected in only 21-23% of primary ovarian cancers but in 81-84% of tumor nodules found on the peritoneal surface at second-look operations following primary chemotherapy. This reflects a 4-fold increase (P<0.0001) in autophagy between primary disease and post-treatment recurrence. We suggest that DIRAS3 not only regulates the AIC, but induces autophagy in dormant, nutrient-deprived ovarian cancer cells that remain after conventional chemotherapy, facilitating their survival.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Maria T Baquero
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Hailing Yang
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Maojie Yang
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Albert S Reger
- Department of Pharmacology; Baylor College of Medicine; Houston, TX USA
| | - Choel Kim
- Department of Pharmacology; Baylor College of Medicine; Houston, TX USA
| | - Douglas A Levine
- Gynecologic Service; Department of Surgery; Memorial Sloan-Kettering Cancer Center; New York, NY USA
| | - Charlotte H Clarke
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Warren S-L Liao
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | - Robert C Bast
- Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| |
Collapse
|
57
|
Structural perspective of ARHI mediated inhibition of STAT3 signaling: An insight into the inactive to active transition of ARHI and its interaction with STAT3 and importinβ. Cell Signal 2015; 27:739-55. [DOI: 10.1016/j.cellsig.2014.11.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/11/2014] [Accepted: 11/21/2014] [Indexed: 01/27/2023]
|
58
|
Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, Kimmelman A, Kumar S, Levine B, Maiuri MC, Martin SJ, Penninger J, Piacentini M, Rubinsztein DC, Simon HU, Simonsen A, Thorburn AM, Velasco G, Ryan KM, Kroemer G. Autophagy in malignant transformation and cancer progression. EMBO J 2015; 34:856-80. [PMID: 25712477 PMCID: PMC4388596 DOI: 10.15252/embj.201490784] [Citation(s) in RCA: 916] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/15/2022] Open
Abstract
Autophagy plays a key role in the maintenance of cellular homeostasis. In healthy cells, such a homeostatic activity constitutes a robust barrier against malignant transformation. Accordingly, many oncoproteins inhibit, and several oncosuppressor proteins promote, autophagy. Moreover, autophagy is required for optimal anticancer immunosurveillance. In neoplastic cells, however, autophagic responses constitute a means to cope with intracellular and environmental stress, thus favoring tumor progression. This implies that at least in some cases, oncogenesis proceeds along with a temporary inhibition of autophagy or a gain of molecular functions that antagonize its oncosuppressive activity. Here, we discuss the differential impact of autophagy on distinct phases of tumorigenesis and the implications of this concept for the use of autophagy modulators in cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Federico Pietrocola
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - José Manuel Bravo-San Pedro
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - Ravi K Amaravadi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark IRCCS Fondazione Santa Lucia and Department of Biology University of Rome Tor Vergata, Rome, Italy
| | - Patrice Codogno
- Université Paris Descartes Sorbonne Paris Cité, Paris, France Institut Necker Enfants-Malades (INEM), Paris, France INSERM U1151, Paris, France CNRS UMR8253, Paris, France
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - David A Gewirtz
- Department of Pharmacology, Toxicology and Medicine, Virginia Commonwealth University, Richmond Virginia, VA, USA
| | | | - Alec Kimmelman
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria Chiara Maiuri
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - Seamus J Martin
- Department of Genetics, Trinity College, The Smurfit Institute, Dublin, Ireland
| | - Josef Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Andrew M Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University of Madrid, Madrid, Spain Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
59
|
|
60
|
Abstract
The WHO grading scheme for glial neoplasms assigns Grade II to 5 distinct tumors of astrocytic or oligodendroglial lineage: diffuse astrocytoma, oligodendroglioma, oligoastrocytoma, pleomorphic xanthoastrocytoma, and pilomyxoid astrocytoma. Although commonly referred to collectively as among the "low-grade gliomas," these 5 tumors represent molecularly and clinically unique entities. Each is the subject of active basic research aimed at developing a more complete understanding of its molecular biology, and the pace of such research continues to accelerate. Additionally, because managing and predicting the course of these tumors has historically proven challenging, translational research regarding Grade II gliomas continues in the hopes of identifying novel molecular features that can better inform diagnostic, prognostic, and therapeutic strategies. Unfortunately, the basic and translational literature regarding the molecular biology of WHO Grade II gliomas remains nebulous. The authors' goal for this review was to present a comprehensive discussion of current knowledge regarding the molecular characteristics of these 5 WHO Grade II tumors on the chromosomal, genomic, and epigenomic levels. Additionally, they discuss the emerging evidence suggesting molecular differences between adult and pediatric Grade II gliomas. Finally, they present an overview of current strategies for using molecular data to classify low-grade gliomas into clinically relevant categories based on tumor biology.
Collapse
Affiliation(s)
- Nicholas F Marko
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | |
Collapse
|
61
|
Loss of ARHI expression in colon cancer and its clinical significance. Contemp Oncol (Pozn) 2014; 18:329-33. [PMID: 25477755 PMCID: PMC4248061 DOI: 10.5114/wo.2014.45764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 11/10/2013] [Accepted: 11/13/2013] [Indexed: 12/28/2022] Open
Abstract
Aim of the study The Ras-related tumour suppressor gene aplasia Ras homolog member I (ARHI) is downregulated in many types of cancer, including ovarian cancer and hepatocellular carcinoma. In the present study, we explore the expression level and role of ARHI in colon cancer. Moreover, the mechanisms that down-regulate expression of ARHI in colon cancer will be further investigated. Material and methods ARHI expression levels were evaluated with immunohistochemistry, reverse transcriptase-PCR, and western blot. Loss of heterozygosity (LOH), single strand conformation polymorphism (SSCP), and methylation-specific PCR (MSP) were used to study the mechanisms of ARHI down-regulation. Results Low expression of ARHI was observed in 61.7% (37/60) of colon cancer specimens. Compared with the paired noncancerous tissues, ARHI expression was significantly decreased in colon cancer tissues. Furthermore, low ARHI expression was significantly associated with worse differentiation degree and Dukes’ stage (P < 0.05). Methylation-specific PCR assay revealed that the methylation rates of ARHI were 53.3% (16/30) and 46.7% (14/30) in ARHI CpG I and CpG II, respectively. Therefore, methylation of promoter may be involving in down regulation of ARHI expression. Conclusions These data highlight an important role for ARHI in colon cancer, which could be a therapeutic strategy against this malignancy.
Collapse
|
62
|
Yu J, Kong CZ, Zhang Z, Zhan B, Jiang ZM. Aplasia Ras homolog member I expression induces apoptosis in renal cancer cells via the β-catenin signaling pathway. Mol Med Rep 2014; 11:475-81. [PMID: 25339197 DOI: 10.3892/mmr.2014.2742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 09/18/2014] [Indexed: 11/06/2022] Open
Abstract
In numerous types of cancer, the Ras-associated tumor suppressor gene aplasia Ras homolog member I (ARHI), is downregulated. However, the function of ARHI in renal cancer remains to be elucidated. The present study investigated whether the suppressor gene ARHI influenced the growth of renal cancer cell lines and aimed to elucidate its mechanism of action, using the techniques of cell biology and molecular pathology. To the best of our knowledge, the present study was the first to determine the effects of ARHI on human renal cancer cells in vivo and in vitro. It was demonstrated that ARHI exhibited a tumor suppressor function in OS-RC-2 cells and acted via the β-catenin signaling pathway. It was additionally confirmed that the levels of ARHI messenger RNA and protein in renal cancer tissues were lower than those in matched normal tissues. These results provided a novel insight into the possible therapeutic applications of ARHI in renal cancer.
Collapse
Affiliation(s)
- Jian Yu
- Department of Urology, Institute of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chui-Ze Kong
- Department of Urology, Institute of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Zhang
- Department of Urology, Institute of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Bo Zhan
- Department of Urology, Institute of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhen-Ming Jiang
- Department of Urology, Institute of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
63
|
JMJD2A-dependent silencing of Sp1 in advanced breast cancer promotes metastasis by downregulation of DIRAS3. Breast Cancer Res Treat 2014; 147:487-500. [PMID: 25193278 DOI: 10.1007/s10549-014-3083-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/26/2014] [Indexed: 12/14/2022]
Abstract
Specificity protein 1(Sp1) is a ubiquitous transcription factor and is highly expressed in breast cancer. However, its expression pattern and role in breast cancer progression remain unclear. The purpose of this study is to examine the expression pattern of Sp1 and determine its role in breast cancer progression. Immunohistochemistry (IHC) was performed on breast cancer tissues to reveal the expression pattern of Sp1. Spearman rank correlation was used for clinical statistics. Gene and protein expressions were monitored by IHC analysis, quantitative polymerase chain reaction, and Western blot. Wound-healing and Transwell assays were conducted to assess the role of Sp1 in breast cancer. Co-immunoprecipitation, deletion mutagenesis, chromatin immunoprecipitation, and dual luciferase reporter gene assays were used for investigation of the regulatory network. Sp1 expression was downregulated in late stage breast cancer and in highly invasive breast cancer cell lines. Expression of Sp1 was negatively correlated with TNM staging (P = 0.002) and metastasis status (P = 0.023). Overexpression of Sp1 inhibited breast cancer cell migratory and invasive abilities, whereas knockdown of GTP-binding RAS-like 3 (DIRAS3, also known as ARHI, NOEY2) attenuated the inhibitory effects. Moreover, re-expression of DIRAS3 abolished Sp1 knockdown-mediated cell migration and invasion. Jumonji domain containing 2A (JMJD2A) inhibited Sp1 autoregulation and explains Sp1 expression pattern in breast cancer. Sp1 negatively regulated breast cancer metastasis by transcriptional activation of DIRAS3. Inhibition of Sp1 autoregulation by JMJD2A contributed to Sp1 expression pattern in breast cancer. Our findings provided evidence that targeted therapy against Sp1 might be useful in early stage breast cancer. However, in late stages, development of Sp1 activator may be more promising for breast cancer treatments.
Collapse
|
64
|
EZH2-Induced H3K27me3 is Associated with Epigenetic Repression of the ARHI Tumor-Suppressor Gene in Ovarian Cancer. Cell Biochem Biophys 2014; 71:105-12. [DOI: 10.1007/s12013-014-0168-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
65
|
An open-pollinated design for mapping imprinting genes in natural populations. Brief Bioinform 2014; 16:449-60. [DOI: 10.1093/bib/bbu019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/14/2014] [Indexed: 02/04/2023] Open
|
66
|
Li LL, Xue AM, Li BX, Shen YW, Li YH, Luo CL, Zhang MC, Jiang JQ, Xu ZD, Xie JH, Zhao ZQ. JMJD2A contributes to breast cancer progression through transcriptional repression of the tumor suppressor ARHI. Breast Cancer Res 2014; 16:R56. [PMID: 24886710 PMCID: PMC4077733 DOI: 10.1186/bcr3667] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 05/22/2014] [Indexed: 12/17/2022] Open
Abstract
Introduction Breast cancer is a worldwide health problem and the leading cause of cancer death among females. We previously identified Jumonji domain containing 2A (JMJD2A) as a critical mediator of breast cancer proliferation, migration and invasion. We now report that JMJD2A could promote breast cancer progression through transcriptional repression of the tumor suppressor aplasia Ras homolog member I (ARHI). Methods Immunohistochemistry was performed to examine protein expressions in 155 cases of breast cancer and 30 non-neoplastic tissues. Spearman correlation analysis was used to analyze the correlation between JMJD2A expression and clinical parameters as well as several tumor regulators in 155 cases of breast cancer. Gene and protein expressions were monitored by quantitative polymerase chain reaction (qPCR) and Western blot. Results from knockdown of JMJD2A, overexpression of JMJD2A, Co-immunoprecipitation (Co-IP) assay, dual luciferase reporter gene assay and chromatin immunoprecipitation (ChIP) elucidated molecular mechanisms of JMJD2A action in breast cancer progression. Furthermore, the effects of ARHI overexpression on JMJD2A-mediated tumor progression were investigated in vitro and in vivo. For in vitro experiments, cell proliferation, wound-healing, migration and invasion were monitored by cell counting, scratch and Boyden Chamber assays. For in vivo experiments, control cells and cells stably expressing JMJD2A alone or together with ARHI were inoculated into mammary fat pads of mice. Tumor volume, tumor weight and metastatic nodules were measured by caliper, electronic balance and nodule counting, respectively. Results JMJD2A was highly expressed in human breast cancers and positively correlated with tumor progression. Knockdown of JMJD2A increased ARHI expression whereas overexpression of JMJD2A decreased ARHI expression at both protein and mRNA levels. Furthermore, E2Fs and histone deacetylases were involved in the transcriptional repression of ARHI expression by JMJD2A. And the aggressive behavior of JMJD2A in breast cancers could be reversed by re-expression of ARHI in vitro and in vivo. Conclusion We demonstrated a cancer-promoting effect of JMJD2A and defined a novel molecular pathway contributing to JMJD2A-mediated breast cancer progression.
Collapse
|
67
|
Lu Z, Yang H, Sutton MN, Yang M, Clarke CH, Liao WSL, Bast RC. ARHI (DIRAS3) induces autophagy in ovarian cancer cells by downregulating the epidermal growth factor receptor, inhibiting PI3K and Ras/MAP signaling and activating the FOXo3a-mediated induction of Rab7. Cell Death Differ 2014; 21:1275-89. [PMID: 24769729 DOI: 10.1038/cdd.2014.48] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 01/31/2023] Open
Abstract
The process of autophagy has been described in detail at the molecular level in normal cells, but less is known of its regulation in cancer cells. Aplasia Ras homolog member I (ARHI; DIRAS3) is an imprinted tumor suppressor gene that is downregulated in multiple malignancies including ovarian cancer. Re-expression of ARHI slows proliferation, inhibits motility, induces autophagy and produces tumor dormancy. Our previous studies have implicated autophagy in the survival of dormant ovarian cancer cells and have shown that ARHI is required for autophagy induced by starvation or rapamycin treatment. Re-expression of ARHI in ovarian cancer cells blocks signaling through the PI3K and Ras/MAP pathways, which, in turn, downregulates mTOR and initiates autophagy. Here we show that ARHI is required for autophagy-meditated cancer cell arrest and ARHI inhibits signaling through PI3K/AKT and Ras/MAP by enhancing internalization and degradation of the epidermal growth factor receptor. ARHI-mediated downregulation of PI3K/AKT and Ras/ERK signaling also decreases phosphorylation of FOXo3a, which sequesters this transcription factor in the nucleus. Nuclear retention of FOXo3a induces ATG4 and MAP-LC3-I, required for maturation of autophagosomes, and also increases the expression of Rab7, required for fusion of autophagosomes with lysosomes. Following the knockdown of FOXo3a or Rab7, autophagolysosome formation was observed but was markedly inhibited, resulting in numerous enlarged autophagosomes. ARHI expression correlates with LC3 expression and FOXo3a nuclear localization in surgical specimens of ovarian cancer. Thus, ARHI contributes to the induction of autophagy through multiple mechanisms in ovarian cancer cells.
Collapse
Affiliation(s)
- Z Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - H Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - M N Sutton
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - M Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - C H Clarke
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - W S-L Liao
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| | - R C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-1439, USA
| |
Collapse
|
68
|
Docherty LE, Rezwan FI, Poole RL, Jagoe H, Lake H, Lockett GA, Arshad H, Wilson DI, Holloway JW, Temple IK, Mackay DJG. Genome-wide DNA methylation analysis of patients with imprinting disorders identifies differentially methylated regions associated with novel candidate imprinted genes. J Med Genet 2014; 51:229-38. [PMID: 24501229 PMCID: PMC3963529 DOI: 10.1136/jmedgenet-2013-102116] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/04/2013] [Accepted: 12/09/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Genomic imprinting is allelic restriction of gene expression potential depending on parent of origin, maintained by epigenetic mechanisms including parent of origin-specific DNA methylation. Among approximately 70 known imprinted genes are some causing disorders affecting growth, metabolism and cancer predisposition. Some imprinting disorder patients have hypomethylation of several imprinted loci (HIL) throughout the genome and may have atypically severe clinical features. Here we used array analysis in HIL patients to define patterns of aberrant methylation throughout the genome. DESIGN We developed a novel informatic pipeline capable of small sample number analysis, and profiled 10 HIL patients with two clinical presentations (Beckwith-Wiedemann syndrome and neonatal diabetes) using the Illumina Infinium Human Methylation450 BeadChip array to identify candidate imprinted regions. We used robust statistical criteria to quantify DNA methylation. RESULTS We detected hypomethylation at known imprinted loci, and 25 further candidate imprinted regions (nine shared between patient groups) including one in the Down syndrome critical region (WRB) and another previously associated with bipolar disorder (PPIEL). Targeted analysis of three candidate regions (NHP2L1, WRB and PPIEL) showed allelic expression, methylation patterns consistent with allelic maternal methylation and frequent hypomethylation among an additional cohort of HIL patients, including six with Silver-Russell syndrome presentations and one with pseudohypoparathyroidism 1B. CONCLUSIONS This study identified novel candidate imprinted genes, revealed remarkable epigenetic convergence among clinically divergent patients, and highlights the potential of epigenomic profiling to expand our understanding of the normal methylome and its disruption in human disease.
Collapse
|
69
|
ZUO XIAOXIAO, QIN YAN, ZHANG XIAOJIN, NING QIAN, SHAO SHAN, LUO MINNA, YUAN NA, HUANG SHANGKE, ZHAO XINHAN. Breast cancer cells are arrested at different phases of the cell cycle following the re-expression of ARHI. Oncol Rep 2014; 31:2358-64. [DOI: 10.3892/or.2014.3107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/12/2014] [Indexed: 11/06/2022] Open
|
70
|
Post-natal imprinting: evidence from marsupials. Heredity (Edinb) 2014; 113:145-55. [PMID: 24595366 DOI: 10.1038/hdy.2014.10] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/19/2013] [Accepted: 01/09/2014] [Indexed: 12/31/2022] Open
Abstract
Genomic imprinting has been identified in therian (eutherian and marsupial) mammals but not in prototherian (monotreme) mammals. Imprinting has an important role in optimising pre-natal nutrition and growth, and most imprinted genes are expressed and imprinted in the placenta and developing fetus. In marsupials, however, the placental attachment is short-lived, and most growth and development occurs post-natally, supported by a changing milk composition tailor-made for each stage of development. Therefore there is a much greater demand on marsupial females during post-natal lactation than during pre-natal placentation, so there may be greater selection for genomic imprinting in the mammary gland than in the short-lived placenta. Recent studies in the tammar wallaby confirm the presence of genomic imprinting in nutrient-regulatory genes in the adult mammary gland. This suggests that imprinting may influence infant post-natal growth via the mammary gland as it does pre-natally via the placenta. Similarly, an increasing number of imprinted genes have been implicated in regulating feeding and nurturing behaviour in both the adult and the developing neonate/offspring in mice. Together these studies provide evidence that genomic imprinting is critical for regulating growth and subsequently the survival of offspring not only pre-natally but also post-natally.
Collapse
|
71
|
Over-expression of ARHI decreases tumor growth, migration, and invasion in human glioma. Med Oncol 2014; 31:846. [DOI: 10.1007/s12032-014-0846-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/15/2014] [Indexed: 01/14/2023]
|
72
|
Molecular targets for epithelial ovarian cancer. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
73
|
Orfanelli T, Jeong J, Doulaveris G, Holcomb K, Witkin S. Involvement of autophagy in cervical, endometrial and ovarian cancer. Int J Cancer 2013; 135:519-28. [DOI: 10.1002/ijc.28524] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/18/2013] [Indexed: 01/06/2023]
Affiliation(s)
- T. Orfanelli
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - J.M. Jeong
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - G. Doulaveris
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - K. Holcomb
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| | - S.S. Witkin
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medical College; New York NY
| |
Collapse
|
74
|
Lyu T, Jia N, Wang J, Yan X, Yu Y, Lu Z, Bast RC, Hua K, Feng W. Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 2013; 8:1330-46. [PMID: 24135786 DOI: 10.4161/epi.26675] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The initiation of angiogenesis can mark the transition from tumor dormancy to active growth and recurrence. Mechanisms that regulate recurrence in human cancers are poorly understood, in part because of the absence of relevant models. The induction of ARHI (DIRAS3) induces dormancy and autophagy in human ovarian cancer xenografts but produces autophagic cell death in culture. The addition of VEGF to cultures maintains the viability of dormant autophagic cancer cells, thereby permitting active growth when ARHI is downregulated, which mimics the "recurrence" of growth in xenografts. Two inducible ovarian cancer cell lines, SKOv3-ARHI and Hey-ARHI, were used. The expression level of angiogenesis factors was evaluated by real-time PCR, immunohistochemistry, immunocytochemistry and western blot; their epigenetic regulation was measured by bisulfite sequencing and chromatin immunoprecipitation. Six of the 15 angiogenesis factors were upregulated in dormant cancer cells (tissue inhibitor of metalloproteinases-3, TIMP3; thrombospondin-1, TSP1; angiopoietin-1; angiopoietin-2; angiopoietin-4; E-cadherin, CDH1). We found that TIMP3 and CDH1 expression was regulated epigenetically and was related inversely to the DNA methylation of their promoters in cell cultures and in xenografts. Increased H3K9 acetylation was associated with higher TIMP3 expression in dormant SKOv3-ARHI cells, while decreased H3K27me3 resulted in the upregulation of TIMP3 in dormant Hey-ARHI cells. Elevated CDH1 expression during dormancy was associated with an increase in both H3K4me3 and H3K9Ac in two cell lines. CpG demethylating agents and/or histone deacetylase inhibitors inhibited the re-growth of dormant cancer cells, which was associated with the re-expression of anti-angiogenic genes. The expression of the anti-angiogenic genes TIMP3 and CDH1 is elevated during dormancy and is reduced during the transition to active growth by changes in DNA methylation and histone modification.
Collapse
Affiliation(s)
- Tianjiao Lyu
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Nan Jia
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Jieyu Wang
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Xiaohui Yan
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Yinhua Yu
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Zhen Lu
- Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Robert C Bast
- Department of Experimental Therapeutics; University of Texas, M.D. Anderson Cancer Center; Houston, TX USA
| | - Keqin Hua
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| | - Weiwei Feng
- Department of Gynecology; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Obstetrics and Gynecology Hospital; Fudan University; Shanghai, PR China
| |
Collapse
|
75
|
Pinho FG, Frampton AE, Nunes J, Krell J, Alshaker H, Jacob J, Pellegrino L, Roca-Alonso L, de Giorgio A, Harding V, Waxman J, Stebbing J, Pchejetski D, Castellano L. Downregulation of microRNA-515-5p by the estrogen receptor modulates sphingosine kinase 1 and breast cancer cell proliferation. Cancer Res 2013; 73:5936-48. [PMID: 23928990 DOI: 10.1158/0008-5472.can-13-0158] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sphingosine kinase 1 (SK1) plays an important role in estrogen-dependent breast tumorigenesis, but its regulation is poorly understood. A subset of microRNAs (miRNA, miR) is regulated by estrogen and contributes to cellular proliferation and cancer progression. Here, we describe that miR-515-5p is transcriptionally repressed by estrogen receptor α (ERα) and functions as a tumor suppressor in breast cancer. Its downregulation enhances cell proliferation and estrogen-dependent SK1 activity, mediated by a reduction of miR-515-5p posttranscriptional repression. Enforced expression of miR-515-5p in breast cancer cells causes a reduction in SK1 activity, reduced cell proliferation, and the induction of caspase-dependent apoptosis. Conversely, opposing effects occur with miR-515-5p inhibition and by SK1 silencing. Notably, we show that estradiol (E2) treatment downregulates miR-515-5p levels, whereas the antiestrogen tamoxifen causes a decrease in SK1, which is rescued by silencing miR-515-5p. Analysis of chromatin immunoprecipitation sequencing (ChIP-Seq) data reveals that miR-515-5p suppression is mediated by a direct interaction of ERα within its promoter. RNA-sequencing (RNA-Seq) analysis of breast cancer cells after overexpressing miR-515-5p indicates that it partly modulates cell proliferation by regulating the Wnt pathway. The clinical implications of this novel regulatory system are shown as miR-515-5p is significantly downregulated in ER-positive (n = 146) compared with ER-negative (n = 98) breast cancers. Overall, we identify a new link between ERα, miR-515-5p, proliferation, and apoptosis in breast cancer tumorigenesis.
Collapse
Affiliation(s)
- Filipa G Pinho
- Authors' Affiliations: Division of Oncology, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM); Department of Medicine and HPB Surgical Unit, Department of Surgery & Cancer, Imperial College; Department of Oncology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London; and School of Medicine, University of East Anglia, Norwich, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
The ARHI (aplasia Ras homologue member I, also known as DIRAS3) gene shows 60.0% sequence homology to the Ras proto-oncogene and was the first mater-nally-imprinted tumor suppressor gene identified in the Ras family. It is constitutively expressed from the paternal allele in normal breast, ovary, heart, liver, pancreas, thyroid and brain tissues, and is lost or markedly down-regulated primarily in breast, ovarian, pancreas and thyroid tumor tissues. We have investigated the expression, LOH (loss of heterozygosity) and methylation status of this gene in glial tumors and peripheral blood samples of 21 patients, and in seven normal brain tissue samples. Gene expression by real time reverse transcriptase polymerase chain reaction (RT-PCR) was found to be increased in 14 and decreased in seven of the 21 tumors. The LOH was detected by fragment analysis, using five labeled polymorphic markers specific for the 1p31 region, in two of the tumors. Methylation status of the CpG island I, II and III was evaluated using COBRA (combined bisulfite restriction analysis) and RFLP (restriction fragment length polymorphism) in 21 tumors and also a hypermethylated healthy volunteer as a positive control, revealed that only two tumors had hypermethylation in CpG island I (of which one also had LOH). These results suggest that LOH and hypermethylation may be one mechanism of silencing the ARHI gene expression and development of glial tumor development.
Collapse
|
77
|
Niemczyk M, Ito Y, Huddleston J, Git A, Abu-Amero S, Caldas C, Moore G, Stojic L, Murrell A. Imprinted chromatin around DIRAS3 regulates alternative splicing of GNG12-AS1, a long noncoding RNA. Am J Hum Genet 2013; 93:224-35. [PMID: 23871723 PMCID: PMC3738830 DOI: 10.1016/j.ajhg.2013.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/19/2013] [Accepted: 06/07/2013] [Indexed: 12/21/2022] Open
Abstract
Imprinted gene clusters are regulated by long noncoding RNAs (lncRNAs), CCCTC binding factor (CTCF)-mediated boundaries, and DNA methylation. DIRAS3 (also known as ARH1 or NOEY1) is an imprinted gene encoding a protein belonging to the RAS superfamily of GTPases and is located within an intron of a lncRNA called GNG12-AS1. In this study, we investigated whether GNG12-AS1 is imprinted and coregulated with DIRAS3. We report that GNG12-AS1 is coexpressed with DIRAS3 in several tissues and coordinately downregulated with DIRAS3 in breast cancers. GNG12-AS1 has several splice variants, all of which initiate from a single transcription start site. In placenta tissue and normal cell lines, GNG12-AS1 is biallelically expressed but some isoforms are allele-specifically spliced. Cohesin plays a role in allele-specific splicing of GNG12-AS1. In breast cancer cell lines with loss of DIRAS3 imprinting, DIRAS3 and GNG12-AS1 are silenced in cis and the remaining GNG12-AS1 transcripts are predominantly monoallelic. The GNG12-AS1 locus, which includes DIRAS3, provides an example of imprinted cotranscriptional splicing and a potential model system for studying the long-range effects of CTCF-cohesin binding on splicing and transcriptional interference.
Collapse
Affiliation(s)
| | - Yoko Ito
- Cancer Research UK, Robinson Way, Cambridge CB2 0RE, UK
| | | | - Anna Git
- Cancer Research UK, Robinson Way, Cambridge CB2 0RE, UK
| | - Sayeda Abu-Amero
- Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Carlos Caldas
- Cancer Research UK, Robinson Way, Cambridge CB2 0RE, UK
| | - Gudrun E. Moore
- Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | | | - Adele Murrell
- Cancer Research UK, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
78
|
Li Y, Liu M, Zhang Y, Han C, You J, Yang J, Cao C, Jiao S. Effects of ARHI on breast cancer cell biological behavior regulated by microRNA-221. Tumour Biol 2013; 34:3545-54. [PMID: 23801152 DOI: 10.1007/s13277-013-0933-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/12/2013] [Indexed: 12/26/2022] Open
Abstract
The aplysia ras homolog member I (ARHI) is a tumor suppressor gene and is downregulated in various cancers. The downregulation of ARHI was regulated by miR-221 in prostate cancer cell lines. However, it has not been reported whether ARHI is regulated by miR-221 in breast cancer. Here, we reported that the ARHI protein level was downregulated in breast cancer tissues and breast cancer cell lines. The overexpression of ARHI could inhibit cell proliferation and invasion and induce cell apoptosis. To address whether ARHI is regulated by miR-221 in breast cancer cell lines, the results in this study showed that a significant inverse correlation existed between ARHI and miR-221. MiR-221 displayed an upregulation in breast cancer tissues and breast cancer cell lines. The inhibition of miR-221 induced a significant upregulation of ARHI in MCF-7 cells. To prove a direct interaction between miR-221 and ARHI mRNA, ARHI 3'UTR, which includes the potential target site for miR-221, was cloned downstream of the luciferase reporter gene of the pMIR-REPORT vector to generate the pMIR-ARHI-3'UTR vector. The results confirmed a direct interaction of miR-221 with a target site on the 3'UTR of ARHI. In conclusion, ARHI is a tumor suppressor gene that is downregulated in breast cancer. The overexpression of ARHI could inhibit breast cancer cell proliferation and invasion and induce cell apoptosis. This study demonstrated for the first time that the downregulation of ARHI in breast cancer cells could be regulated by miR-221.
Collapse
Affiliation(s)
- Ying Li
- Department of Oncology, Chinese PLA General Hospital, No. 28, FuXing Road, Beijing, 100853, China
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Li J, Cui G, Sun L, Wang SJ, Li YL, Meng YG, Guan Z, Fan WS, Li LA, Yang YZ, You YQ, Fu XY, Yan ZF, Huang K. STAT3 acetylation-induced promoter methylation is associated with downregulation of the ARHI tumor-suppressor gene in ovarian cancer. Oncol Rep 2013; 30:165-70. [PMID: 23604529 DOI: 10.3892/or.2013.2414] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/02/2013] [Indexed: 11/06/2022] Open
Abstract
ARHI is a Ras-related imprinted tumor-suppressor gene that inhibits cancer cell growth and motility. ARHI is downregulated in the majority of ovarian cancer cells, and promoter methylation is considered to be associated with its loss of expression. however, the underlying mechanisms are not well understood. Thus, the present study aimed to investigate the specific functions of ARHI and its methylation in ovarian cancer cell proliferation. Furthermore, we examined the possible role of acetylated STAT3 in modulating the expression of ARHI and its methylation. In accordance with the majority of previous studies, reduced ARHI expression was found in epithelial ovarian cancer tissues and cancer cell lines as indicated by immunohistochemistry and RT-PCR. In addition, CpG islands I and II within ARHI promoter regions were partially methylated or hypermethylated in cancer cell lines (SKOV-3 and HO-8910) as analyzed by pyrosequencing assays, resulting in enhanced proliferation of the cancer cells. This proliferation was reversed by the administration of 5-aza-2'-deoxycytidine. Subsequently, we demonstrated that STAT3 acetylation was increased in HO-8910 cells, and the methylation status of CpG I was altered in response to the acetylation of STAT3 using western blotting. Finally, chromatin immunoprecipitation (ChIP) and IP analysis indicated that acetylated STAT3 bound to the ARHI promoter and recruited DNA methyltransferase 1 for genetic modification. In conclusion, acetylated STAT3-induced promoter gene methylation accounts for the loss of ARHI expression and cancer cell proliferation.
Collapse
Affiliation(s)
- Jie Li
- Department of Gynecology and Obstetrics, General Hospital of PLA, Beijing 100853, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Hu YQ, Si LJ, Ye ZS, Lin ZH, Zhou JP. Inhibitory effect of ARHI on pancreatic cancer cells and NF-κB activity. Mol Med Rep 2013; 7:1180-4. [PMID: 23447002 DOI: 10.3892/mmr.2013.1342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 02/19/2013] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the effect of aplasia ras homolog member I (ARHI) on proliferation, apoptosis and the cell cycle in the pancreatic cancer cell line PANC-1. The study also aimed to examine the effect of ARHI on the activity of the nuclear factor (NF)-κB and to determine whether ARHI acts as a tumor suppressor in the development of pancreatic cancer by inhibiting the activity of NF-κB. A pIRES2‑EGFP‑ARHI vector, constructed by reverse transcrition (RT)‑PCR, was transiently transfected into the PANC-1 cells and analyzed for the expression of the ARHI protein by western blotting. A MTT assay was used to quantify cell proliferation, and apoptosis was analyzed by flow cytometry. The NF‑κB signaling pathway, specifically the pathway using the nuclear phosphorylated p65 isoform, was analyzed by western blotting. Expression of the ARHI protein was detected by western blotting subsequent to the PANC-1 cells being transiently transfected with the pIRES2‑EGFP‑ARHI construct. Cell proliferation was strongly inhibited in the PANC-1 cells transfected with pIRES2‑EGFP‑ARHI. The cell cycle assays indicated an increase in the number of cells at the G0/G1 phase and a decrease in the cells at the S phase, but the difference was not significant (P>0.05). Time course studies also indicated a marked increase in the apoptotic index following transient transfection, as well as a gradual decrease in the expression of the nuclear phosphorylated p65 protein. ARHI acts as a tumor suppressor by downregulating the NF‑κB signaling pathway, which results in the inhibition of cell proliferation, apoptosis and the cell cycle in the pancreatic tumor PANC-1 cell line.
Collapse
Affiliation(s)
- Yi-Qun Hu
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian 361004, P.R. China.
| | | | | | | | | |
Collapse
|
81
|
Zhang S, Feng XL, Shi L, Gong CJ, He ZJ, Wu HJ, Ling TY. Genome-wide analysis of DNA methylation in tongue squamous cell carcinoma. Oncol Rep 2013; 29:1819-26. [PMID: 23446731 DOI: 10.3892/or.2013.2309] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 01/23/2013] [Indexed: 11/06/2022] Open
Abstract
Tongue squamous cell carcinoma (TSCC) is one of the most common types of oral cancer; however, its molecular mechanisms remain unclear. In this study, methylated DNA immunoprecipitation (MeDIP) coupled with methylation microarray analysis was performed to screen for aberrantly methylated genes in adjacent normal control and TSCC tissues from 9 patients. Roche NimbleGen Human DNA Methylation 385K Promoter Plus CpG Island Arrays were used to detect 28,226 CpG sites. A total of 1,269 hypermethylated CpG sites covering 330 genes and 1,385 hypomethylated CpG sites covering 321 genes were found in TSCC tissue, compared to the adjacent normal tissue. Furthermore, we chose three candidate genes (FBLN1, ITIH5 and RUNX3) and validated the DNA methylation status by methylation-specific PCR (MS-PCR) and the mRNA expression levels by reverse transcription PCR (RT-PCR). In TSCC tissue, FBLN1 and ITIH5 were shown to be hypermethylated and their expression was found to be decreased, and RUNX3 was shown to be hypomethylated, however, its mRNA expression was found to be increased. In addition, another three genes (BCL2L14, CDCP1 and DIRAS3) were tested by RT-PCR. In TSCC tissue, BCL2L14 and CDCP1 expressions were markedly upregulated, and DIRAS3 expression was significantly downregulated. Our data demonstrated that aberrant DNA methylation is observed in TSCC tissue and plays an important role in the tumorigenesis, development and progression of TSCC.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Oral and Maxillofacial Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China.
| | | | | | | | | | | | | |
Collapse
|
82
|
Zhu YH, Fu L, Chen L, Qin YR, Liu H, Xie F, Zeng T, Dong SS, Li J, Li Y, Dai Y, Xie D, Guan XY. Downregulation of the novel tumor suppressor DIRAS1 predicts poor prognosis in esophageal squamous cell carcinoma. Cancer Res 2013; 73:2298-309. [PMID: 23436800 DOI: 10.1158/0008-5472.can-12-2663] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Loss of chromosome 19p is one of the most frequent allelic imbalances in esophageal squamous cell carcinoma (ESCC), suggesting the existence of one or more tumor suppressor genes within this region. In this study, we investigated a role in ESCCs for a candidate tumor suppressor gene located at 19p13.3, the Ras-like small GTPase DIRAS1. Downregulation of DIRAS1 occurred in approximately 50% of primary ESCCs where it was associated significantly with advanced clinical stage, lymph node metastasis, and poor overall survival. LOH and promoter methylation analyses suggested that loss of DIRAS1 expression was mediated by epigenetic mechanisms. Functional studies established that ectopic re-expression of DIRAS1 in ESCC cells inhibited cell proliferation, clonogenicity, cell motility, and tumor formation. Mechanistic investigations suggested that DIRAS1 acted through extracellular signal-regulated kinase (ERK1/2; MAPK3/1) and p38 mitogen-activated protein kinase (MAPK; MAPK14) signaling to trigger BAD Ser112 dephosphorylation and matrix metalloproteinase (MMP)2/9 transcriptional inactivation to promote apoptosis and inhibit metastasis, respectively. Taken together, our results revealed that DIRAS1 has a pivotal function in ESCC pathogenesis, with possible use as a biomarker and intervention point for new therapeutic strategies.
Collapse
Affiliation(s)
- Ying-Hui Zhu
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Court F, Martin-Trujillo A, Romanelli V, Garin I, Iglesias-Platas I, Salafsky I, Guitart M, Perez de Nanclares G, Lapunzina P, Monk D. Genome-wide allelic methylation analysis reveals disease-specific susceptibility to multiple methylation defects in imprinting syndromes. Hum Mutat 2013; 34:595-602. [PMID: 23335487 DOI: 10.1002/humu.22276] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/07/2013] [Indexed: 12/12/2022]
Abstract
Genomic imprinting is the parent-of-origin-specific allelic transcriptional silencing observed in mammals, which is governed by DNA methylation established in the gametes and maintained throughout the development. The frequency and extent of epimutations associated with the nine reported imprinting syndromes varies because it is evident that aberrant preimplantation maintenance of imprinted differentially methylated regions (DMRs) may affect multiple loci. Using a custom Illumina GoldenGate array targeting 27 imprinted DMRs, we profiled allelic methylation in 65 imprinting defect patients. We identify multilocus hypomethylation in numerous Beckwith-Wiedemann syndrome, transient neonatal diabetes mellitus (TNDM), and pseudohypoparathyroidism 1B patients, and an individual with Silver-Russell syndrome. Our data reveal a broad range of epimutations exist in certain imprinting syndromes, with the exception of Prader-Willi syndrome and Angelman syndrome patients that are associated with solitary SNRPN-DMR defects. A mutation analysis identified a 1 bp deletion in the ZFP57 gene in a TNDM patient with methylation defects at multiple maternal DMRs. In addition, we observe missense variants in ZFP57, NLRP2, and NLRP7 that are not consistent with maternal effect and aberrant establishment or methylation maintenance, and are likely benign. This work illustrates that further extensive molecular characterization of these rare patients is required to fully understand the mechanism underlying the etiology of imprint establishment and maintenance.
Collapse
Affiliation(s)
- Franck Court
- Imprinting and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Jia L, Jin H, Zhou J, Chen L, Lu Y, Ming Y, Yu Y. A potential anti-tumor herbal medicine, Corilagin, inhibits ovarian cancer cell growth through blocking the TGF-β signaling pathways. Altern Ther Health Med 2013; 13:33. [PMID: 23410205 PMCID: PMC3598193 DOI: 10.1186/1472-6882-13-33] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 02/11/2013] [Indexed: 11/21/2022]
Abstract
Background Phyllanthus niruri L. is a well-known hepatoprotective and antiviral medicinal herb. Recently, we identified Corilagin as a major active component with anti-tumor activity in this herbal medicine. Corilagin is a member of the tannin family that has been discovered in many medicinal plants and has been used as an anti-inflammatory agent. However, there have been few reports of the anti-tumor effects of Corilagin, and its anti-tumor mechanism has not been investigated clearly. The aim of the present study is to investigate the anticancer properties of Corilagin in ovarian cancer cells. Methods The ovarian cancer cell lines SKOv3ip, Hey and HO-8910PM were treated with Corilagin and analyzed by Sulforhodamine B (SRB) cell proliferation assay, flow cytometry, and reverse phase protein array (RPPA). Corilagin was delivered intraperitoneally to mice bearing SKOv3ip xenografts. Results Corilagin inhibited the growth of the ovarian cancer cell lines SKOv3ip and Hey, with IC50 values of less than 30 μM, while displaying low toxicity against normal ovarian surface epithelium cells, with IC50 values of approximately 160 μM. Corilagin induced cell cycle arrest at the G2/M stage and enhanced apoptosis in ovarian cancer cells. Immunoblotting assays demonstrated that Cyclin B1, Myt1, Phospho-cdc2 and Phospho-Weel were down-regulated after Corilagin treatment. Xenograft tumor growth was significantly lower in the Corilagin-treated group compared with the untreated control group (P <0.05). More interestingly, Corilagin inhibited TGF-β secretion into the culture supernatant of all tested ovarian cancer cell lines and blocked the TGF-β-induced stabilization of Snail. In contrast, a reduction of TGF-β secretion was not observed in cancer cells treated with the cytotoxic drug Paclitaxel, suggesting that Corilagin specifically targets TGF-β secretion. Corilagin blocked the activation of both the canonical Smad and non-canonical ERK/AKT pathways. Conclusions Corilagin extracted from Phyllanthus niruri L. acts as a natural, effective therapeutic agent against the growth of ovarian cancer cells via targeted action against the TGF-β/AKT/ERK/Smad signaling pathways.
Collapse
|
85
|
Klingauf M, Beck M, Berge U, Turgay Y, Heinzer S, Horvath P, Kroschewski R. The tumour suppressor DiRas3 interacts with C-RAF and downregulates MEK activity to restrict cell migration. Biol Cell 2012; 105:91-107. [DOI: 10.1111/boc.201200030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 11/13/2012] [Indexed: 11/26/2022]
|
86
|
Yang H, Lu X, Qian J, Xu F, Hu Y, Yu Y, Bast RC, Li J. Imprinted tumor suppressor gene ARHI induces apoptosis correlated with changes in DNA methylation in pancreatic cancer cells. Mol Med Rep 2012; 3:581-7. [PMID: 21472283 DOI: 10.3892/mmr_00000301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aplesia Ras homologue member I (ARHI, DIRAS3) is a Ras-related imprinted growth inhibitory gene whose expression is down-regulated in the majority of breast and ovarian cancers. This study investigated the inhibitory function of ARHI in pancreatic cancer. Six pancreatic cancer cell lines, tumor xenografts in nude mice and 20 pancreatic cancer tissue sections were analyzed. ARHI is widely expressed in ductal and acinar cells of normal pancreatic tissue, but is down-regulated or lost in approximately 50% of pancreatic cancers. Aberrant methylation of the ARHI locus was found in five pancreatic cancer cell lines, which exhibited down-regulation or loss of ARHI expression. Hypermethylation was detected in five cell lines (5/5, 100%) at CpG island I, in two cell lines (2/5, 40%) at CpG island II and in four cell lines (4/5, 80%) at CpG island III. Re-expression of ARHI significantly inhibited the growth of pancreatic cancer cells. This inhibition was associated with the induction of apoptosis. Treatment with the demethylating agent 5-aza-2'deoxycytidine (5-aza-dC) restored ARHI mRNA expression, inhibited cell growth and induced apoptosis in PANC-1 and P3 human pancreatic cancer cells in culture. In nu/nu mice, 5-aza-dC also inhibited the growth of PANC-1 xenografts and induced apoptosis, as observed by TUNEL staining. These effects were associated with the re-expression of ARHI protein. Therefore, ARHI may serve as a growth inhibitory gene in a significant fraction of pancreatic cancers. Re-expression of ARHI significantly induced the apoptosis of pancreatic cancer cells. A demethylation agent reduced human pancreatic cancer cell line growth in conjunction with ARHI re-expression.
Collapse
Affiliation(s)
- Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Li BX, Luo CL, Li H, Yang P, Zhang MC, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ. Effects of siRNA-mediated knockdown of jumonji domain containing 2A on proliferation, migration and invasion of the human breast cancer cell line MCF-7. Exp Ther Med 2012; 4:755-761. [PMID: 23170139 PMCID: PMC3501409 DOI: 10.3892/etm.2012.662] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/30/2012] [Indexed: 12/16/2022] Open
Abstract
Jumonji domain containing 2A (JMJD2A) is a potential cancer-associated gene that may be involved in human breast cancer. The present study aimed to investigate suppressive effects on the MCF-7 human breast cancer cell line by transfection with JMJD2A-specific siRNA. Quantitative real-time PCR and western blot analysis were used to detect the expression levels of JMJD2A. Flow cytometric (FCM) analysis and WST-8 assay were used to evaluate cell proliferation. Boyden chambers were used in cell migration and invasion assays to evaluate the cell exercise capacity. Expression levels of JMJD2A mRNA and protein in the siRNA group were both downregulated successfully by transfection. FCM results showed that the percentage of cells in the G0/G1 phase in the siRNA group was significantly greater than that in the blank (P<0.05) and negative control groups (P<0.05). Additionally, the mean absorbance in the siRNA group was significantly lower (P<0.05), as observed by WST-8 assay. Moreover, a decreased number of migrated cells in the siRNA group was observed (P<0.05) using a cell migration and invasion assay. These data indicated that knockdown of JMJD2A may cause inhibition of proliferation, migration and invasion of MCF-7 cells. This study provides a new perspective in understanding the molecular mechanisms underlying the progression of breast cancer and offers a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Bei-Xu Li
- Department of Forensic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Tang HL, Hu YQ, Qin XP, Jazag A, Yang H, Yang YX, Yang XN, Liu JJ, Chen JM, Guleng B, Ren JL. Aplasia ras homolog member I is downregulated in gastric cancer and silencing its expression promotes cell growth in vitro. J Gastroenterol Hepatol 2012; 27:1395-404. [PMID: 22497484 DOI: 10.1111/j.1440-1746.2012.07146.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Aplasia ras homolog member I (ARHI) is a maternally imprinted tumor suppressor gene. ARHI protein is widely expressed in many types of human tissues; however, its expression is frequently reduced or absent in various tumors and plays a tumor suppressor role for in vitro study. In this study, we investigated the expression level of ARHI in gastric cancer in order to investigate the function of ARHI and signaling pathways that might be linked during gastric cancer development. METHODS ARHI mRNA and protein expression levels were analyzed in primary gastric cancer tissues, adjacent noncancerous gastric tissues and gastric cancer cell lines using semi-quantitative polymerase chain reaction, western blotting and immunohistochemistry, respectively. RESULTS Our results showed that both mRNA and protein expression levels of the ARHI gene were significantly downregulated (P < 0.05) in gastric cancer tissues and cell lines compared to the corresponding normal control groups. The protein expression level of ARHI was not associated with age, gender, location of tumor, tumor size or metastasis in patients with gastric cancer. However, a significant correlation between the level of ARHI protein expression and the degree of tumor differentiation and Tumor-Node-Metastasis stage was observed (P < 0.05). Furthermore, results of the methyl thiazolyl tetrazolium and Transwell assays and flow cytometric analysis showed increased cell proliferation, migration and anti-apoptotic capacities in the well-differentiated gastric cancer MKN-28 cell line, which has stably silenced ARHI protein expression. CONCLUSION Our data indicate that ARHI expression is downregulated in human gastric cancer and it may be a novel tumor suppressive target for gastric cancer therapy.
Collapse
Affiliation(s)
- Hai-Ling Tang
- Department of Gastroenterology, Zhongshan Hospital affiliated with Xiamen University, Fujian Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Baljuls A, Beck M, Oenel A, Robubi A, Kroschewski R, Hekman M, Rudel T, Rapp UR. The tumor suppressor DiRas3 forms a complex with H-Ras and C-RAF proteins and regulates localization, dimerization, and kinase activity of C-RAF. J Biol Chem 2012; 287:23128-40. [PMID: 22605333 DOI: 10.1074/jbc.m112.343780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The maternally imprinted Ras-related tumor suppressor gene DiRas3 is lost or down-regulated in more than 60% of ovarian and breast cancers. The anti-tumorigenic effect of DiRas3 is achieved through several mechanisms, including inhibition of cell proliferation, motility, and invasion, as well as induction of apoptosis and autophagy. Re-expression of DiRas3 in cancer cells interferes with the signaling through Ras/MAPK and PI3K. Despite intensive research, the mode of interference of DiRas3 with the Ras/RAF/MEK/ERK signal transduction is still a matter of speculation. In this study, we show that DiRas3 associates with the H-Ras oncogene and that activation of H-Ras enforces this interaction. Furthermore, while associated with DiRas3, H-Ras is able to bind to its effector protein C-RAF. The resulting multimeric complex consisting of DiRas3, C-RAF, and active H-Ras is more stable than the two protein complexes H-Ras·C-RAF or H-Ras·DiRas3, respectively. The consequence of this complex formation is a DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity.
Collapse
Affiliation(s)
- Angela Baljuls
- Theodor Boveri Institute of Bioscience, Department of Microbiology, University of Wuerzburg, 97074 Wuerzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Li Y, Shi L, Han C, Wang Y, Yang J, Cao C, Jiao S. Effects of ARHI on cell cycle progression and apoptosis levels of breast cancer cells. Tumour Biol 2012; 33:1403-10. [PMID: 22528939 DOI: 10.1007/s13277-012-0388-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 03/21/2012] [Indexed: 12/12/2022] Open
Abstract
The purposes of this study were to investigate the role of Aplysia Ras Homolog I (ARHI) on cell growth, proliferation, apoptosis, and other biological characteristics of HER2-positive breast cancer cells. Our goal was to provide experimental evidence for the development of future effective treatments of HER2-positive breast cancer. A pcDNA3.1-ARHI eukaryotic expression vector was constructed and transfected into the human HER2-positive breast cancer cell lines SK-BR-3 and JIMT-1. Then, various experimental methods were utilized to analyze the biological characteristics of ARHI-expressing breast cancer cells and to examine the impact of expression of the ARHI gene on cyclin D1, p27(Kip1), and calpain1 expression. We further analyzed the cells in each group after treatment with trastuzumab to examine the effects of this drug on various cellular characteristics. When we compared pcDNA3.1-ARHI-expressing SK-BR-3 and JIMT-1 cells to their respective empty vector and control groups, we found that cell viability was significantly lower (p < 0.05) in the ARHI-expressing cells, and the proportions of G1 phase cells and apoptotic cells were significantly higher in the ARHI-expressing cells (p < 0.05). In all groups of SK-BR-3 cells, trastuzumab treatment significantly decreased cell growth (p < 0.05). The proportion of cells in G1 phase and the number of apoptotic cells in the pcDNA3.1-ARHI-expressing group were significantly higher than that in the empty vector group and the control group (p < 0.05). The growth of pcDNA3.1-ARHI-transfected JIMT-1 cells was significantly decreased (p < 0.05), while the proportion of apoptotic cells was significantly increased (p < 0.05). Cell growth, viability, and the percentage of apoptotic cells were similar between the JIMT-1 empty vector and control groups. ARHI expression inhibited cyclin D1 expression in SK-BR-3 cells and JIMT-1 cells, while it promoted p27(Kip1) and calpain1 expression in these cells. ARHI expression inhibits the growth and proliferation of HER2-positive breast cancer cells, while it also promotes apoptosis in these cells. ARHI expression also improves the sensitivity of JIMT-1 cells to trastuzumab by inducing apoptosis.
Collapse
Affiliation(s)
- Ying Li
- Department of Oncology, Chinese PLA General Hospital, No. 28, FuXing Road, Beijing, 100853, China
| | | | | | | | | | | | | |
Collapse
|
91
|
Lin D, Cui F, Bu Q, Yan C. The expression and clinical significance of GTP-binding RAS-like 3 (ARHI) and microRNA 221 and 222 in prostate cancer. J Int Med Res 2012; 39:1870-5. [PMID: 22117988 DOI: 10.1177/147323001103900530] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Globally, prostate cancer is the most common malignancy among men and there is no biomarker for defining tumour invasion and progression. Guanosine-5'-triphosphate (GTP)-binding RAS-like 3 (ARHI) is a tumour suppressor gene that has been found to be downregulated in the prostate cancer cell line PC-3. MicroRNA 221 and 222 have been shown to regulate ARHI expression negatively. This study evaluated tissue samples from patients with prostate cancer (n = 35) that were designated as aggressive or non-aggressive according to their Gleason grade. Expression of ARHI and microRNA 221 and 222 was measured by real-time reverse transcription-polymerase chain reaction. The level of ARHI mRNA was significantly lower in aggressive compared with non-aggressive prostate cancer tissue samples. In contrast, microRNA 221 and 222 levels were significantly higher in aggressive compared with non-aggressive prostate cancer tissue samples. Whether ARHI and microRNA 221 and 222 could be considered as biomarkers for disease progression in prostate cancer requires further investigation.
Collapse
Affiliation(s)
- D Lin
- Urology Department, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | | | | | | |
Collapse
|
92
|
Grossi V, Simone C. Special Agents Hunting Down Women Silent Killer: The Emerging Role of the p38α Kinase. JOURNAL OF ONCOLOGY 2012; 2012:382159. [PMID: 22481926 PMCID: PMC3317177 DOI: 10.1155/2012/382159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/21/2011] [Accepted: 12/29/2011] [Indexed: 02/08/2023]
Abstract
Ovarian cancer is sensitive to chemotherapy with platinum compounds; however, the therapy success rate is significantly lowered by a high incidence of recurrence and by the acquisition of drug resistance. These negative outcomes mainly depend on altered apoptotic and drug resistance pathways, determining the need for the design of new therapeutic strategies to improve patient survival. This challenge has become even more critical because it has been recognized that hindering uncontrolled cell growth is not sufficient as the only curative approach. In fact, while current therapies are mostly conceived to impair survival of highly proliferating cells, several lines of research are now focusing on cancer-specific features to specifically target malignant cells with the aim of avoiding drug resistance and reducing adverse effects. Recently, great interest has been generated by the identification of metabolic reprogramming mechanisms occurring in cancer cells, such as the increase in glycolysis levels. In this light, pharmacologic manipulation of relevant pathways involved in cancer-specific metabolism and drug resistance could prove an effective approach to treat ovarian cancer patients.
Collapse
Affiliation(s)
| | - Cristiano Simone
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology (DTP), Consorzio Mario NegriSud 66030, Santa Maria Imbaro, Italy
| |
Collapse
|
93
|
Bast RC. Molecular approaches to personalizing management of ovarian cancer. Ann Oncol 2012; 22 Suppl 8:viii5-viii15. [PMID: 22180401 DOI: 10.1093/annonc/mdr516] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cytoreductive surgery and empirical combination chemotherapy have improved 5-year survival for ovarian cancer patients, but have not increased the overall rate of cure. Poor outcomes relate, at least in part, to late diagnosis and to the persistence of dormant ovarian cancer cells that have resisted conventional drugs. Increased understanding of the molecular, cellular and clinical biology of ovarian cancer must be translated into personalized therapy with conventional and targeted agents as well as personalized detection of high-grade cancers in early stages. Different strategies will be required to treat low-grade and high-grade serous cancers as well as other histotypes. Activating mutations of Ras and Raf can be targeted in low-grade cancers. Activation of the PI3K pathway-PI3Kness-and inactivation of BRCA function-BRCAness-can be targeted in high-grade lesions. Inhibition of multiple pathways will be required. Sensitivity of primary cancers to paclitaxel and platinum can be modulated by inhibiting kinases and other molecules that regulate the cell cycle. Dormant ovarian cancer cells may depend upon autophagy, cytokines and growth factors for survival. Early detection can utilize two stage strategies where rising serum biomarker levels prompt imaging in a small fraction of women. Screening can be personalized by taking into account each woman's baseline biomarker levels.
Collapse
Affiliation(s)
- R C Bast
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA.
| |
Collapse
|
94
|
Engler DA, Gupta S, Growdon WB, Drapkin RI, Nitta M, Sergent PA, Allred SF, Gross J, Deavers MT, Kuo WL, Karlan BY, Rueda BR, Orsulic S, Gershenson DM, Birrer MJ, Gray JW, Mohapatra G. Genome wide DNA copy number analysis of serous type ovarian carcinomas identifies genetic markers predictive of clinical outcome. PLoS One 2012; 7:e30996. [PMID: 22355333 PMCID: PMC3280266 DOI: 10.1371/journal.pone.0030996] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 12/28/2011] [Indexed: 01/09/2023] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer death in women. Ovarian cancers display a high degree of complex genetic alterations involving many oncogenes and tumor suppressor genes. Analysis of the association between genetic alterations and clinical endpoints such as survival will lead to improved patient management via genetic stratification of patients into clinically relevant subgroups. In this study, we aim to define subgroups of high-grade serous ovarian carcinomas that differ with respect to prognosis and overall survival. Genome-wide DNA copy number alterations (CNAs) were measured in 72 clinically annotated, high-grade serous tumors using high-resolution oligonucleotide arrays. Two clinically annotated, independent cohorts were used for validation. Unsupervised hierarchical clustering of copy number data derived from the 72 patient cohort resulted in two clusters with significant difference in progression free survival (PFS) and a marginal difference in overall survival (OS). GISTIC analysis of the two clusters identified altered regions unique to each cluster. Supervised clustering of two independent large cohorts of high-grade serous tumors using the classification scheme derived from the two initial clusters validated our results and identified 8 genomic regions that are distinctly different among the subgroups. These 8 regions map to 8p21.3, 8p23.2, 12p12.1, 17p11.2, 17p12, 19q12, 20q11.21 and 20q13.12; and harbor potential oncogenes and tumor suppressor genes that are likely to be involved in the pathogenesis of ovarian carcinoma. We have identified a set of genetic alterations that could be used for stratification of high-grade serous tumors into clinically relevant treatment subgroups.
Collapse
Affiliation(s)
- David A. Engler
- Department of Statistics, Brigham Young University, Provo, Utah, United States of America
| | - Sumeet Gupta
- Whitehead Institute of Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Whitfield B. Growdon
- Department of Vincent Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Ronny I. Drapkin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Mai Nitta
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Petra A. Sergent
- Department of Vincent Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Serena F. Allred
- Department of Statistics, Brigham Young University, Provo, Utah, United States of America
| | - Jenny Gross
- Women's Cancer Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Michael T. Deavers
- Department of Pathology and Gynecology Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Wen-Lin Kuo
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Beth Y. Karlan
- Women's Cancer Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bo R. Rueda
- Department of Vincent Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Sandra Orsulic
- Women's Cancer Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - David M. Gershenson
- Department of Pathology and Gynecology Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael J. Birrer
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Joe W. Gray
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Gayatry Mohapatra
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
95
|
Abstract
Breast cancer is caused by progressive genetic alterations and epigenetic changes that trigger tumor development. Among the epigenetic alterations descried in breast cancer, DNA promoter methylation has been extensively studied and observed in genes involved in several critical signaling pathways that initiate and promote breast tumorigenesis. In addition to DNA hypermethylation, global methylation levels have been observed to decrease with breast cancer progression. In close connection with DNA methylation changes, histone modifications, especially the acetylation and methylation of histone lysines, play an essential role in the nucleosomes remodeling and gene expression regulation in breast cancer.
Collapse
Affiliation(s)
- Ramona G Dumitrescu
- Saba University School of Medicine, Saba Island and Georgetown University Medical School, Washington, DC, USA.
| |
Collapse
|
96
|
Li BX, Zhang MC, Luo CL, Yang P, Li H, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ. Effects of RNA interference-mediated gene silencing of JMJD2A on human breast cancer cell line MDA-MB-231 in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:90. [PMID: 21962223 PMCID: PMC3215938 DOI: 10.1186/1756-9966-30-90] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 10/03/2011] [Indexed: 11/24/2022]
Abstract
Previous data demonstrate that JMJD2A is a cancer-associated gene and may be involved in human breast cancer by demethylation of H3K9me3. The aim of this study was to investigate depressive effects on JMJD2A by transfection with JMJD2A-sepcific siRNA in human breast cancer cell line MDA-MB-231 and effects on cell proliferation, invasion and migration. JMJD2A-specific siRNA was chemically synthesised and transfected into human breast cancer cell line MDA-MB-231. Expression levels of JMJD2A were detected by quantitative real-time PCR and Western blot analysis. Cells proliferation was evaluated by using flow cytometric anlysis and MTT assay. The abilities of invasion and migration were evaluated by cell migration and invasion assay with Boyden chambers. The results showed that the transfection was successful and expression levels of JMJD2A mRNA and protein in siRNA group were both down-regulated. By MTT assay, the mean actual absorbance in siRNA group was significantly lower than that in blank control group (P < 0.05) and negative control group (P < 0.05). In addition, the percentage of cells in G0/G1 phase in siRNA group was significantly more than that in blank control group (P < 0.05) and negative control group (P < 0.05). Furthermore, by cell invasion and migration assay, the decreased number of migrated cells in siRNA group was observed (P < 0.05). These data imply that silencing JMJD2A gene could result in cell cycle change and proliferation inhibition, and lead to suppress tumor cell invasion and migration. It provides a new perspective in understanding the pleiotropic functions of JMJD2A and its contribution to human breast cancer.
Collapse
Affiliation(s)
- Bei-Xu Li
- Department of Forensic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
A nucleolar protein, H19 opposite tumor suppressor (HOTS), is a tumor growth inhibitor encoded by a human imprinted H19 antisense transcript. Proc Natl Acad Sci U S A 2011; 108:16759-64. [PMID: 21940503 DOI: 10.1073/pnas.1110904108] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The H19 gene, which localizes within a chromosomal region on human chromosome 11p15 that is commonly lost in Wilms tumor (WT), encodes an imprinted untranslated RNA. However, the biological significance of the H19 noncoding transcript remains unresolved because replacement of the RNA transcript with a neocassette has no obvious phenotypic effect. Here we show that the human H19 locus also encodes a maternally expressed, translated gene, antisense to the known H19 transcript, which is conserved in primates. This gene, termed HOTS for H19 opposite tumor suppressor, encodes a protein that localizes to the nucleus and nucleolus and that interacts with the human enhancer of rudimentary homolog (ERH) protein. WTs that show loss of heterozygosity of 11p15 or loss of imprinting of IGF2 also silence HOTS (7/7 and 10/10, respectively). Overexpression of HOTS inhibits Wilms, rhabdoid, rhabdomyosarcoma, and choriocarcinoma tumor cell growth, and silencing HOTS by RNAi increases in vitro colony formation and in vivo tumor growth. These results demonstrate that the human H19 locus harbors an imprinted gene encoding a tumor suppressor protein within the long-sought WT2 locus.
Collapse
|
98
|
Berardi DE, Campodónico PB, Díaz Bessone MI, Urtreger AJ, Todaro LB. Autophagy: friend or foe in breast cancer development, progression, and treatment. Int J Breast Cancer 2011; 2011:595092. [PMID: 22295229 PMCID: PMC3262577 DOI: 10.4061/2011/595092] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/11/2011] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a catabolic process responsible for the degradation and recycling of long-lived proteins and organelles by lysosomes. This degradative pathway sustains cell survival during nutrient deprivation, but in some circumstances, autophagy leads to cell death. Thereby, autophagy can serve as tumor suppressor, as the reduction in autophagic capacity causes malignant transformation and spontaneous tumors. On the other hand, this process also functions as a protective cell-survival mechanism against environmental stress causing resistance to antineoplastic therapies. Although autophagy inhibition, combined with anticancer agents, could be therapeutically beneficial in some cases, autophagy induction by itself could lead to cell death in some apoptosis-resistant cancers, indicating that autophagy induction may also be used as a therapy. This paper summarizes the most important findings described in the literature about autophagy and also discusses the importance of this process in clinical settings.
Collapse
Affiliation(s)
- Damian E Berardi
- Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, C1417DTB Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
99
|
Aplasia Ras homologue member I overexpression induces apoptosis through inhibition of survival pathways in human hepatocellular carcinoma cells in culture and in xenograft. Cell Biol Int 2011; 35:1019-24. [DOI: 10.1042/cbi20110023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
100
|
Taneja P, Zhu S, Maglic D, Fry EA, Kendig RD, Inoue K. Transgenic and knockout mice models to reveal the functions of tumor suppressor genes. Clin Med Insights Oncol 2011; 5:235-57. [PMID: 21836819 PMCID: PMC3153120 DOI: 10.4137/cmo.s7516] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cancer is caused by multiple genetic alterations leading to uncontrolled cell proliferation through multiple pathways. Malignant cells arise from a variety of genetic factors, such as mutations in tumor suppressor genes (TSGs) that are involved in regulating the cell cycle, apoptosis, or cell differentiation, or maintenance of genomic integrity. Tumor suppressor mouse models are the most frequently used animal models in cancer research. The anti-tumorigenic functions of TSGs, and their role in development and differentiation, and inhibition of oncogenes are discussed. In this review, we summarize some of the important transgenic and knockout mouse models for TSGs, including Rb, p53, Ink4a/Arf, Brca1/2, and their related genes.
Collapse
Affiliation(s)
| | - Sinan Zhu
- The Departments of Pathology
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Dejan Maglic
- The Departments of Pathology
- Cancer Biology
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | - Kazushi Inoue
- The Departments of Pathology
- Cancer Biology
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|