201
|
Liu R, Wang L, Chen G, Katoh H, Chen C, Liu Y, Zheng P. FOXP3 up-regulates p21 expression by site-specific inhibition of histone deacetylase 2/histone deacetylase 4 association to the locus. Cancer Res 2009; 69:2252-9. [PMID: 19276356 PMCID: PMC2715174 DOI: 10.1158/0008-5472.can-08-3717] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
p21 loss has been implicated in conferring oncogenic activity to known tumor suppressor gene KLF4 and cancer drug tamoxifen. Regulators of p21, therefore, play critical roles in tumorigenesis. Here, we report that X-linked tumor suppressor FOXP3 is essential for p21 expression in normal epithelia and that lack of FOXP3 is associated with p21 down-regulation in breast cancer samples. A specific FOXP3 binding site in the intron 1 is essential for p21 induction by FOXP3. FOXP3 specifically inhibited binding of histone deacetylase 2 (HDAC2) and HDAC4 to the site and increased local histone H3 acetylation. Short hairpin RNA silencing of either HDAC2 or HDAC4 is sufficient to induce p21 expression. Our data provides a novel mechanism for transcription activation by FOXP3 and a genetic mechanism for lack of p21 in a large proportion of breast cancer.
Collapse
Affiliation(s)
- Runhua Liu
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Lizhong Wang
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Guoyun Chen
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Hiroto Katoh
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Chong Chen
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Yang Liu
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Pan Zheng
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
- Department of Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| |
Collapse
|
202
|
Sangodkar J, Shi J, DiFeo A, Schwartz R, Bromberg R, Choudhri A, McClinch K, Hatami R, Scheer E, Kremer-Tal S, Martignetti JA, Hui A, Leung WK, Friedman SL, Narla G. Functional role of the KLF6 tumour suppressor gene in gastric cancer. Eur J Cancer 2009; 45:666-76. [PMID: 19101139 PMCID: PMC2970616 DOI: 10.1016/j.ejca.2008.11.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 10/31/2008] [Accepted: 11/05/2008] [Indexed: 11/17/2022]
Abstract
Gastric cancer is the second most common cancer and a leading cause of cancer-related death worldwide. The Kruppel-like factor 6 (KLF6) tumour suppressor gene had been previously shown to be inactivated in a number of human cancers through loss of heterozygosity (LOH), somatic mutation, decreased expression and increased alternative splicing into a dominant negative oncogenic splice variant, KLF6-SV1. In the present study, 37 gastric cancer samples were analysed for the presence of loss of heterozygosity (LOH) of the KLF6 locus and somatic mutation. In total, 18 of 34 (53%) of the gastric cancer samples analysed demonstrated KLF6 locus specific loss. Four missense mutations, such as T179I, R198G, R71Q and S180L, were detected. Interestingly, two of these mutations R71Q and S180L have been identified independently by several groups in various malignancies including prostate, colorectal and gastric cancers. In addition, decreased wild-type KLF6 (wtKLF6) expression was associated with loss of the KLF6 locus and was present in 48% of primary gastric tumour samples analysed. Functional studies confirmed that wtKLF6 suppressed proliferation of gastric cancer cells via transcriptional regulation of the cyclin-dependent kinase inhibitor p21 and the oncogene c-myc. Functional characterisation of the common tumour-derived mutants demonstrated that the mutant proteins fail to suppress proliferation and function as dominant negative regulators of wtKLF6 function. Furthermore, stable overexpression of the R71Q and S180L tumour-derived mutants in the gastric cancer cell line, Hs746T, resulted in an increased tumourigenicity in vivo. Combined, these findings suggest an important role for the KLF6 tumour suppressor gene in gastric cancer development and progression and identify several highly cancer-relevant signalling pathways regulated by the KLF6 tumour suppressor gene.
Collapse
Affiliation(s)
- Jaya Sangodkar
- Department of Medicine, Mount Sinai School of Medicine, New York, NY
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Jiayan Shi
- Department of Medicine, Mount Sinai School of Medicine, New York, NY
| | - Analisa DiFeo
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Rachel Schwartz
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Romina Bromberg
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Aisha Choudhri
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Kim McClinch
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Raheleh Hatami
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Elias Scheer
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - Sigal Kremer-Tal
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| | - John A Martignetti
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY
- Department of Pediatrics, Mount Sinai School of Medicine, New York, NY
| | - Alex Hui
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong
| | - WK Leung
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong
| | - Scott L Friedman
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY
| | - Goutham Narla
- Department of Medicine, Mount Sinai School of Medicine, New York, NY
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
203
|
Telerman A, Amson R. The molecular programme of tumour reversion: the steps beyond malignant transformation. Nat Rev Cancer 2009; 9:206-16. [PMID: 19180095 DOI: 10.1038/nrc2589] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
How cells become malignant has preoccupied scientists for over a century. However, the converse question is also valid: are tumour cells capable of reverting from their malignant state? Askanazy's studies in 1907 indicated that teratoma cells could differentiate into normal somatic tissues and current evidence indicates that some tumour cells have acquired the molecular circuitry that results in the negation of chromosomal instability, translocations, oncogene activation and loss of tumour suppressor genes. Studying these extremely rare events of tumour reversion and deciphering these pathways, which involve SIAH1, presenilin 1, TSAP6 and translationally controlled tumour protein (TCTP), could lead to new avenues in cancer treatment.
Collapse
Affiliation(s)
- Adam Telerman
- LBPA, UMR 8113, Ecole Normale Supérieure, 61 Avenue du Président Wilson, 94235 Cachan, France.
| | | |
Collapse
|
204
|
DiFeo A, Martignetti JA, Narla G. The role of KLF6 and its splice variants in cancer therapy. Drug Resist Updat 2008; 12:1-7. [PMID: 19097929 DOI: 10.1016/j.drup.2008.11.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 11/03/2008] [Accepted: 11/04/2008] [Indexed: 12/21/2022]
Abstract
The Krüppel-like zinc finger transcription factor (KLF6) gene encodes a family of proteins generated through alternative splicing involved in the regulation of cancer development and progression. Alternative splicing of the KLF6 gene results in the production of at least four alternatively spliced isoforms, two of which are extensively discussed in this review. The full length form of the KLF6 gene is a tumor suppressor gene that is frequently inactivated by loss of heterozygozity (LOH), somatic mutation, and/or decreased expression in human cancer. While the exact mechanisms underlying KLF6's tumor suppressor roles are not completely known, a number of highly relevant, overlapping pathways have been described: transactivation of p21 in a p53-independent manner, reduction of cyclin D1/cdk4 complexes via interaction with cyclin D1, inhibition of c-Jun proto-oncoprotein activities, decreased VEGF expression, and induction of apoptosis. Kruppel-like factor 6 splice variant 1 (KLF6-SV1) is an oncogenic splice variant of the KLF6 tumor suppressor gene that is specifically overexpressed in a number of human cancers. Increased KLF6-SV1 expression is associated with poor prognosis in prostate, lung, and ovarian cancer. Furthermore, KLF6-SV1 has been shown to be biologically active, antagonizing the tumor suppressor function of KLF6 and promoting tumor growth and dissemination in both ovarian and prostate cancer models. In addition, a common germline polymorphism in the KLF6 gene associated with increased prostate cancer risk in a large multi-institutional study of 3411 men results in increased expression of KLF6-SV1. Furthermore, recent studies have demonstrated that targeted reduction of KLF6-SV1 results in the induction of spontaneous apoptosis in cell culture, synergizes with chemotherapeutic agents like cisplatin, and results in significant tumor regression in vivo. Combined, these data make the KLF6 gene family a compelling therapeutic target for both the treatment of localized as well as metastatic cancer.
Collapse
Affiliation(s)
- Analisa DiFeo
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
205
|
Sirintrapun SJ, Parwani AV. Molecular Pathology of the Genitourinary Tract: Prostate and Bladder. Surg Pathol Clin 2008; 1:211-36. [PMID: 26837907 DOI: 10.1016/j.path.2008.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The knowledge of cellular mechanisms in tumors of the prostate and bladder has grown exponentially. Molecular technologies have led to the discovery of TMPRSS2 in prostate cancer and the molecular pathways distinguishing low- and high-grade urothelial neoplasms. UroVysion with fluorescence in situ hybridization is already commonplace as an adjunct to cytologic diagnosis of urothelial neoplasms. This trend portends the future in which classification and diagnosis of tumors of the prostate and bladder through morphologic analysis will be supplemented by molecular information correlating with prognosis and targeted therapy. This article outlines tumor molecular pathology of the prostate and bladder encompassing current genomic, epigenomic, and proteonomic findings.
Collapse
Affiliation(s)
- S Joseph Sirintrapun
- Pathology Informatics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh Medical Center Shadyside Hospital, Room WG 07, 5230 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
206
|
KLF6 and TP53 mutations are a rare event in prostate cancer: distinguishing between Taq polymerase artifacts and true mutations. Mod Pathol 2008; 21:1470-8. [PMID: 19020536 DOI: 10.1038/modpathol.2008.145] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Krüppel-like factor 6 (KLF6) has been reported to act as a tumor suppressor gene involved in the regulation of the cell cycle by activating p21 in a p53-independent manner. Many studies suggest that KLF6 is inactivated by allelic loss and somatic mutation. However, there is a high variability in the reported frequency of mutations (from 1 to 55%). TP53 also regulates the cell cycle through the activation of p21. In prostate cancer, the reported frequency of TP53 mutations ranges from 3 to 42%. In all these reports, there is a considerable degree of methodological heterogeneity. Our aim was to determine the frequency of KLF6 and TP53 mutations in a well-defined group of prostate tumors with different stages and Gleason grades. The four exons of KLF6 and exons 4-9 of TP53 were studied in 103 cases, including 90 formalin-fixed, paraffin-embedded (FFPE) and 13 frozen samples. All tumors were analyzed through PCR and direct sequencing. All changes found were confirmed by a second independent PCR and sequencing reaction. For KLF6, mutation (E227G) was only detected in one tumor (1%) and for TP53, three different mutations (L130H, H214R, and Y234C) were detected in five tumors (5%). This low mutation index is in keeping with recent papers on the subject. Our study strongly supports the notion that KLF6 and TP53 mutations are not frequent events in prostate cancer. When using FFPE tissues, it is mandatory to perform at least two independent rounds of PCR and sequencing to confirm mutations and exclude Taq polymerase-induced artifacts.
Collapse
|
207
|
Tripathi M, Nandana S, Yamashita H, Ganesan R, Kirchhofer D, Quaranta V. Laminin-332 is a substrate for hepsin, a protease associated with prostate cancer progression. J Biol Chem 2008; 283:30576-84. [PMID: 18784072 PMCID: PMC2576550 DOI: 10.1074/jbc.m802312200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 09/03/2008] [Indexed: 11/06/2022] Open
Abstract
Hepsin, a cell surface protease, is widely reported to be overexpressed in more than 90% of human prostate tumors. Hepsin expression correlates with tumor progression, making it a significant marker and target for prostate cancer. Recently, it was reported that in a prostate cancer mouse model, hepsin up-regulation in tumor tissue promotes progression and metastasis. The underlying mechanisms, however, remain largely uncharacterized. Hepsin transgenic mice displayed reduced laminin-332 (Ln-332) expression in prostate tumors. This is an intriguing cue, since proteolytic processing of extracellular matrix macromolecules, such as Ln-332, is believed to be involved in cancer progression, and Ln-332 expression is lost during human prostate cancer progression. In this study, we provide the first direct evidence that hepsin cleaves Ln-332. Cleavage is specific, since it is both inhibited in a dose-dependent manner by a hepsin inhibitor (Kunitz domain-1) and does not occur when catalytically inactive hepsin is used. By Western blotting and mass spectrometry, we determined that hepsin cleaves the beta3 chain of Ln-332. N-terminal sequencing identified the cleavage site at beta3 Arg(245), in a sequence context (SQLR(245) LQGSCFC) conserved among species and in remarkable agreement with reported consensus target sequences for hepsin activity. In vitro cell migration assays showed that hepsin-cleaved Ln-332 enhanced motility of DU145 prostate cancer cells, which was inhibited by Kunitz domain-1. Further, hepsin-overexpressing LNCaP prostate cancer cells also exhibited increased migration on Ln-332. Direct cleavage of Ln-332 may be one mechanism by which hepsin promotes prostate tumor progression and metastasis, possibly by up-regulating prostate cancer cell motility.
Collapse
Affiliation(s)
- Manisha Tripathi
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
208
|
Koy S, Plaschke J, Luksch H, Friedrich K, Kuhlisch E, Eckelt U, Martinez R. Microsatellite instability and loss of heterozygosity in squamous cell carcinoma of the head and neck. Head Neck 2008; 30:1105-13. [PMID: 18615731 DOI: 10.1002/hed.20857] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Microsatellite instability (MSI) in head and neck squamous cell carcinoma (HNSCC) has been reported with a wide range of frequencies. The aim of our study was to disclose the frequency and basis of MSI in HNSCC and to correlate MSI and findings on loss of heterozygosity (LOH) with the clinical data. METHODS We analyzed MSI and LOH in 91 tumors. All tumors presenting instability were analyzed for the expression of mismatch repair genes (MMR) proteins. RESULTS Low-level microsatellite instability (MSI-L) was seen in 7.7% of the HNSCC. None of the MSI-L tumors had aberrant MMR protein expression. LOH rates up to 57% were identified for different regions on chromosome 3p. For the marker D10S197, we found a significant correlation between LOH and tumor stage IV. CONCLUSION Our results indicate that MMR gene inactivation is rare among primary HNSCC. In contrast, the MSI-L phenotype plays a role in a small subset of tumors. LOH on chromosome arm 3p and 10p12 seems to be involved in tumorigenesis and progression HNSCC, respectively.
Collapse
Affiliation(s)
- Susanne Koy
- Department of Oral and Maxillofacial Surgery, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany.
| | | | | | | | | | | | | |
Collapse
|
209
|
Narla G, DiFeo A, Fernandez Y, Dhanasekaran S, Huang F, Sangodkar J, Hod E, Leake D, Friedman SL, Hall SJ, Chinnaiyan AM, Gerald WL, Rubin MA, Martignetti JA. KLF6-SV1 overexpression accelerates human and mouse prostate cancer progression and metastasis. J Clin Invest 2008; 118:2711-21. [PMID: 18596922 DOI: 10.1172/jci34780] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Accepted: 04/23/2008] [Indexed: 11/17/2022] Open
Abstract
Metastatic prostate cancer (PCa) is one of the leading causes of death from cancer in men. The molecular mechanisms underlying the transition from localized tumor to hormone-refractory metastatic PCa remain largely unknown, and their identification is key for predicting prognosis and targeted therapy. Here we demonstrated that increased expression of a splice variant of the Kruppel-like factor 6 (KLF6) tumor suppressor gene, known as KLF6-SV1, in tumors from men after prostatectomy predicted markedly poorer survival and disease recurrence profiles. Analysis of tumor samples revealed that KLF6-SV1 levels were specifically upregulated in hormone-refractory metastatic PCa. In 2 complementary mouse models of metastatic PCa, KLF6-SV1-overexpressing PCa cells were shown by in vivo and ex vivo bioluminescent imaging to metastasize more rapidly and to disseminate to lymph nodes, bone, and brain more often. Interestingly, while KLF6-SV1 overexpression increased metastasis, it did not affect localized tumor growth. KLF6-SV1 inhibition using RNAi induced spontaneous apoptosis in cultured PCa cell lines and suppressed tumor growth in mice. Together, these findings demonstrate that KLF6-SV1 expression levels in PCa tumors at the time of diagnosis can predict the metastatic behavior of the tumor; thus, KLF-SV1 may represent a novel therapeutic target.
Collapse
Affiliation(s)
- Goutham Narla
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Mihci E, Lindor NM. Germline duplication of chromosomes 10p15.3 and Yp11.32 in a man with learning disability and early onset cutaneous malignant melanoma. Am J Med Genet A 2008; 146A:2298-300. [DOI: 10.1002/ajmg.a.32442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
211
|
Huang X, Li X, Guo B. KLF6 induces apoptosis in prostate cancer cells through up-regulation of ATF3. J Biol Chem 2008; 283:29795-801. [PMID: 18755691 DOI: 10.1074/jbc.m802515200] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
KLF6 (Kruppel-like factor 6) is a zinc finger transcription factor and a tumor suppressor that is frequently mutated in prostate cancer. KLF6 suppresses tumor growth and induces apoptosis in cancer cells through mechanisms still not defined. Here we show that KLF6 induces apoptosis in prostate cancer cells by ATF3 (activating transcription factor 3) expression. KLF6 binds directly to and activates the ATF3 promoter. ATF3 induced apoptosis when ectopically expressed in cells, whereas knockdown of ATF3 by small interference RNA blocked KLF6-induced apoptosis. KLF6 mutants derived from clinical prostate cancers failed to activate the ATF3 promoter and were unable to induce apoptosis. Furthermore, stress conditions (exposure to staurosporine and hypoxia induced by sodium azide) caused significant increase in ATF3 expression and induced apoptosis, whereas knockdown of KLF6 by small interference RNA blocked the increase of ATF3 as well as the induction of apoptosis in these conditions. Thus, ATF3 is a key mediator of KLF6-induced apoptosis in prostate cancer cells.
Collapse
Affiliation(s)
- Xiangwei Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, North Dakota State University, Fargo, North Dakota 58105, USA
| | | | | |
Collapse
|
212
|
Holian J, Qi W, Kelly DJ, Zhang Y, Mreich E, Pollock CA, Chen XM. Role of Kruppel-like factor 6 in transforming growth factor-beta1-induced epithelial-mesenchymal transition of proximal tubule cells. Am J Physiol Renal Physiol 2008; 295:F1388-96. [PMID: 18753303 DOI: 10.1152/ajprenal.00055.2008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Krüppel-like factor 6 (KLF6) is a DNA-binding protein containing a triple zinc-fingered motif and plays a key role in the regulation of cell proliferation, differentiation, and development. More recently it has been implicated in hepatic fibrosis via its binding to the transforming growth factor (TGF)-beta control element. In the kidney, epithelial-mesenchymal transition (EMT) is a major contributor to the pathogenesis of renal fibrosis, with TGF-beta1 being a key mediator of EMT. The present study aimed to determine the role of KLF6 and TGF-beta1 in EMT in proximal tubule cells. To determine the relevance in clinical disease, KLF6 was measured in kidneys of streptozotocin-induced diabetic Ren-2 rats and in cells exposed to high (30 mM) glucose. TGF-beta1 was confirmed to induce EMT by morphological change, loss of E-cadherin, and gain in vimentin expression. KLF6 mRNA expression was concomitantly measured. To determine the role of KLF6 in EMT, the above markers of EMT were determined in KLF6-silenced (small interfering RNA) and KLF6-overexpressing proximal tubule cells. KLF6 overexpression significantly promoted a phenotype consistent with EMT. High glucose induced KLF6 in proximal tubule cells (P < 0.05). This increase in KLF6 in response to high glucose was TGF-beta1 mediated. In an in vivo model of diabetic nephropathy KLF6 increased at week 8 (P < 0.05). KLF6 plays a permissive role in TGF-beta1-induced EMT in proximal tubule cells. Its upregulation in in vivo models of diabetic nephropathy suggests it as a potential therapeutic target.
Collapse
Affiliation(s)
- John Holian
- Kolling Institute, Department of Medicine, Royal North Shore Hospital, Sydney
| | | | | | | | | | | | | |
Collapse
|
213
|
Haining WN, Ebert BL, Subrmanian A, Wherry EJ, Eichbaum Q, Evans JW, Mak R, Rivoli S, Pretz J, Angelosanto J, Smutko JS, Walker BD, Kaech SM, Ahmed R, Nadler LM, Golub TR. Identification of an evolutionarily conserved transcriptional signature of CD8 memory differentiation that is shared by T and B cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:1859-68. [PMID: 18641323 DOI: 10.4049/jimmunol.181.3.1859] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
After Ag encounter, naive lymphocytes differentiate into populations of memory cells that share a common set of functions including faster response to Ag re-exposure and the ability to self-renew. However, memory lymphocytes in different lymphocyte lineages are functionally and phenotypically diverse. It is not known whether discrete populations of T and B cells use similar transcriptional programs during differentiation into the memory state. We used cross-species genomic analysis to examine the pattern of genes up-regulated during the differentiation of naive lymphocytes into memory cells in multiple populations of human CD4, CD8, and B cell lymphocytes as well as two mouse models of memory development. We identified and validated a signature of genes that was up-regulated in memory cells compared with naive cells in both human and mouse CD8 memory differentiation, suggesting marked evolutionary conservation of this transcriptional program. Surprisingly, this conserved CD8 differentiation signature was also up-regulated during memory differentiation in CD4 and B cell lineages. To validate the biologic significance of this signature, we showed that alterations in this signature of genes could distinguish between functional and exhausted CD8 T cells from a mouse model of chronic viral infection. Finally, we generated genome-wide microarray data from tetramer-sorted human T cells and showed profound differences in this differentiation signature between T cells specific for HIV and those specific for influenza. Thus, our data suggest that in addition to lineage-specific differentiation programs, T and B lymphocytes use a common transcriptional program during memory development that is disrupted in chronic viral infection.
Collapse
Affiliation(s)
- W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Hartel M, Narla G, Wente MN, Giese NA, Martignoni ME, Martignetti JA, Friess H, Friedman SL. Increased alternative splicing of the KLF6 tumour suppressor gene correlates with prognosis and tumour grade in patients with pancreatic cancer. Eur J Cancer 2008; 44:1895-903. [PMID: 18691883 DOI: 10.1016/j.ejca.2008.06.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 06/09/2008] [Accepted: 06/16/2008] [Indexed: 01/13/2023]
Abstract
The aim of this study was to correlate the status of the KLF6 tumour suppressor gene including loss of heterozygosity (LOH), mutation and alternative splicing in human pancreatic cancer with tumour grade and survival. Whereas neither KLF6 loss nor mutation was identified, expression of the KLF6 alternative splice forms was significantly increased in pancreatic tumour samples and cell lines. These cancers demonstrated marked cytoplasmic KLF6 expression, consistent with over-expression and accumulation of KLF6 splice form(s), which lack a nuclear localisation signal. In addition, KLF6 splicing correlated significantly with tumour stage and survival. In summary, pancreatic cancer displays a novel pattern of KLF6 dysregulation through selectively increased expression of KLF6 splice variants. Therefore, determination of KLF6 mRNA splicing levels may represent a novel biomarker predicting prognosis.
Collapse
Affiliation(s)
- Mark Hartel
- Department of Surgery, University of Heidelberg, Germany; Department of Surgery, Technische Universität München, Munich, Ismaninger Strasse 22, D-81675 Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
215
|
Miele L, Beale G, Patman G, Nobili V, Leathart J, Grieco A, Abate M, Friedman SL, Narla G, Bugianesi E, Day CP, Reeves HL. The Kruppel-like factor 6 genotype is associated with fibrosis in nonalcoholic fatty liver disease. Gastroenterology 2008; 135:282-291.e1. [PMID: 18515091 PMCID: PMC2891245 DOI: 10.1053/j.gastro.2008.04.004] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 03/17/2008] [Accepted: 04/03/2008] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Although nonalcoholic fatty liver disease (NAFLD) is increasingly common, only a minority of affected individuals develop fibrotic liver disease. Based on its role in liver growth and repair, we explored whether Kruppel-like factor 6 (KLF6) plays a role in NAFLD progression. METHODS KLF6 expression in 31 fibrosis scored NAFLD liver biopsy specimens was assessed by real-time polymerase chain reaction. Transfected minigene constructs were used to study the effect of a polymorphism, KLF6-IVS1-27G>A, that promotes KLF6 alternative splicing in vitro. We genotyped KLF6-IVS1-27G>A in 3 groups of patients (UK group 1, n = 306; Italian group 2, n = 109; trio group 3, n = 61 children and parents). RESULTS KLF6 expression was increased in association with increased steatosis, inflammation, and fibrosis in NAFLD livers. KLF6-IVS1-27G>A promoted alternative splicing of KLF6 and abrogated the up-regulation of both alpha-smooth muscle actin and collagen 1 in LX-2 cells. Group 1 genotyping identified KLF6-IVS1-27G>A in 44 of 306 (14.4%) patients. Notably, KLF6-IVS1-27G>A was associated significantly with milder NAFLD, with only 25% having more advanced fibrosis compared with 45% of wild-type (wt) individuals. This trend was confirmed in group 2. A linear regression analysis including all 415 patients, adjusted for age, sex, body mass index, and blood glucose level, confirmed that presence of the wt KLF6 allele was an independent predictor of fibrotic NAFLD. Furthermore, we have shown preferential transmission of the wt allele to children with fibrotic NAFLD. CONCLUSIONS We report a functional polymorphism in the KLF6 gene associated with advanced NAFLD and believe further study of KLF6 may enhance our understanding of this disease process.
Collapse
Affiliation(s)
- Luca Miele
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Reynolds MA. Molecular alterations in prostate cancer. Cancer Lett 2008; 271:13-24. [PMID: 18554779 DOI: 10.1016/j.canlet.2008.04.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 12/21/2007] [Accepted: 04/24/2008] [Indexed: 01/07/2023]
Abstract
Prostate tumors display a range of clinical phenotypes, from indolent to aggressively metastatic. Numerous gene expression profiling studies have been conducted toward the potential molecular staging of these pathologies, however the identification of genetic markers that predict aggressive disease has not yet been demonstrated in the clinical setting. A recent survey of the literature has shown that molecular alterations in prostate carcinomas can occur through a variety of different mechanisms, ranging from upstream epigenetic changes and genetic polymorphisms to downstream modulations through alternative splicing and other post-translational processes, some of which could involve noncoding RNAs. A summary of these results and recommendations for future work are the subject of this review.
Collapse
|
217
|
Britschgi A, Trinh E, Rizzi M, Jenal M, Ress A, Tobler A, Fey MF, Helin K, Tschan MP. DAPK2 is a novel E2F1/KLF6 target gene involved in their proapoptotic function. Oncogene 2008; 27:5706-16. [PMID: 18521079 DOI: 10.1038/onc.2008.179] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Death-associated protein kinase 2 (DAPK2) belongs to a family of proapoptotic Ca(2+)/calmodulin-regulated serine/threonine kinases. We recently identified DAPK2 as an enhancing factor during granulocytic differentiation. To identify transcriptional DAPK2 regulators, we cloned 2.7 kb of the 5'-flanking region of the DAPK2 gene. We found that E2F1 and Krüppel-like factor 6 (KLF6) strongly activate the DAPK2 promoter. We mapped the E2F1 and KLF6 responsive elements to a GC-rich region 5' of exon 1 containing several binding sites for KLF6 and Sp1 but not for E2F. Moreover, we showed that transcriptional activation of DAPK2 by E2F1 and KLF6 is dependent on Sp1 using Sp1/KLF6-deficient insect cells, mithramycin A treatment to block Sp1-binding or Sp1 knockdown cells. Chromatin immunoprecipitation revealed recruitment of Sp1 and to lesser extent that of E2F1 and KLF6 to the DAPK2 promoter. Activation of E2F1 in osteosarcoma cells led to an increase of endogenous DAPK2 paralleled by cell death. Inhibition of DAPK2 expression resulted in significantly reduced cell death upon E2F1 activation. Similarly, KLF6 expression in H1299 cells increased DAPK2 levels accompanied by cell death that is markedly decreased upon DAPK2 knockdown. Moreover, E2F1 and KLF6 show cooperation in activating the DAPK2 promoter. In summary, our findings establish DAPK2 as a novel Sp1-dependent target gene for E2F1 and KLF6 in cell death response.
Collapse
Affiliation(s)
- A Britschgi
- 1Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
The integrity of genomic DNA is challenged by genotoxic stress originating during normal cellular metabolism or by external insults. Cellular responses to DNA damage involve elegant checkpoint cascades enforcing cell cycle arrest, damage repair, apoptosis or cellular senescence. The loss or alterations of genes involved in the damage response pathways have been reported in many cancer susceptibility syndromes and in sporadic tumors. Furthermore, this surveillance pathway is activated during early tumourigenesis presumably due to uncontrolled replicative cycles and has been recognized as one of the main barriers against the development of cancer. This review discusses the relevance of prostatic epithelial cells in prostate tumourigenesis and highlights common molecular changes associated with prostate cancer. Furthermore, DNA damage responses of primary cultures of human prostatic epithelial cells and fresh human prostate tissues are discussed providing evidence for alterations in crucial DNA damage checkpoint molecules. New insights connecting prostate tumourigenesis to alterations and defects in the pathways maintaining genomic integrity will be discussed.
Collapse
|
219
|
Yea S, Narla G, Zhao X, Garg R, Tal-Kremer S, Hod E, Villanueva A, Loke J, Tarocchi M, Akita K, Shirasawa S, Sasazuki T, Martignetti JA, Llovet JM, Friedman SL. Ras promotes growth by alternative splicing-mediated inactivation of the KLF6 tumor suppressor in hepatocellular carcinoma. Gastroenterology 2008; 134:1521-31. [PMID: 18471523 PMCID: PMC2600656 DOI: 10.1053/j.gastro.2008.02.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 02/02/2008] [Accepted: 02/07/2008] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer worldwide and the third most lethal. Dysregulation of alternative splicing underlies a number of human diseases, yet its contribution to liver cancer has not been explored fully. The Krüppel-like factor 6 (KLF6) gene is a zinc finger transcription factor that inhibits cellular growth in part by transcriptional activation of p21. KLF6 function is abrogated in human cancers owing to increased alternative splicing that yields a dominant-negative isoform, KLF6 splice variant 1 (SV1), which antagonizes full-length KLF6-mediated growth suppression. The molecular basis for stimulation of KLF6 splicing is unknown. METHODS In human HCC samples and cell lines, we functionally link oncogenic Ras signaling to increased alternative splicing of KLF6 through signaling by phosphatidylinositol-3 kinase and Akt, mediated by the splice regulatory protein ASF/SF2. RESULTS In 67 human HCCs, there is a significant correlation between activated Ras signaling and increased KLF6 alternative splicing. In cultured cells, Ras signaling increases the expression of KLF6 SV1, relative to full-length KLF6, thereby enhancing proliferation. Abrogation of oncogenic Ras signaling by small interfering RNA (siRNA) or a farnesyl-transferase inhibitor decreases KLF6 SV1 and suppresses growth. Growth inhibition by farnesyl-transferase inhibitor in transformed cell lines is overcome by ectopic expression of KLF6 SV1. Down-regulation of the splice factor ASF/SF2 by siRNA increases KLF6 SV1 messenger RNA levels. KLF6 alternative splicing is not coupled to its transcriptional regulation. CONCLUSIONS Our findings expand the role of Ras in human HCC by identifying a novel mechanism of tumor-suppressor inactivation through increased alternative splicing mediated by an oncogenic signaling cascade.
Collapse
Affiliation(s)
- Steven Yea
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Goutham Narla
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, 10029
| | - Xiao Zhao
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Rakhi Garg
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Sigal Tal-Kremer
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, 10029
| | - Eldad Hod
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Augusto Villanueva
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Johnny Loke
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Mirko Tarocchi
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Kunihara Akita
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| | - Senji Shirasawa
- Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Takehiko Sasazuki
- Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - John A Martignetti
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, 10029
| | - Josep M Llovet
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
- BCLC Group, Liver Unit, Hospital Clinic, Barcelona
| | - Scott L Friedman
- Division of Liver Diseases and Department of Medicine, New York, NY, 10029
| |
Collapse
|
220
|
Abstract
The Krüppel-like factor (KLF) proteins are zinc finger-containing transcription factors that exert important functions in regulating diverse biologic processes such as growth, proliferation, differentiation, development, inflammation, and apoptosis. Many KLFs have also been shown to play significant roles in tumorigenesis of various organs and tissues. Three in particular-KLF4, KLF5, and KLF6-are often dysregulated in tumors of the gastrointestinal tract, including colorectal cancer. This article reviews the functions of these three KLFs in normal gastrointestinal biology and their pathobiologic roles in colorectal cancer.
Collapse
|
221
|
Bentov I, Narla G, Schayek H, Akita K, Plymate SR, LeRoith D, Friedman SL, Werner H. Insulin-like growth factor-i regulates Kruppel-like factor-6 gene expression in a p53-dependent manner. Endocrinology 2008; 149:1890-7. [PMID: 18174288 DOI: 10.1210/en.2007-0844] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
High-circulating IGF-I concentrations are associated with an increased risk for breast, prostate, and colorectal cancer. Krüppel-like factor-6 (KLF6) is a zinc finger tumor suppressor inactivated in prostate and other types of cancer. We have previously demonstrated that KLF6 is a potent transactivator of the IGF-I receptor promoter. The aim of the present study was to examine the potential regulation of KLF6 gene expression by IGF-I. The human colon cancer cell lines HCT116 +/+ and -/- (with normal and disrupted p53, respectively) were treated with IGF-I. Western blots, quantitative RT-PCR, and transfection assays were used to evaluate the effect of IGF-I on KLF-6 production. Signaling pathway inhibitors were used to identify the mechanisms responsible for regulation of KLF6 expression. Small interfering RNA against p53 and KLF6 was used to assess the role of p53 in regulation of KLF6 expression by IGF-I and to evaluate KLF6 involvement in cell cycle control. Results obtained showed that IGF-I stimulated KLF-6 transcription in cells with normal, but not disrupted, p53, suggesting that KLF6 is a downstream target for IGF-I action. Stimulation of KLF6 expression by IGF-I in a p53-dependent manner may constitute a novel mechanism of action of IGF-I, with implications in normal cell cycle progression and cancer biology.
Collapse
Affiliation(s)
- Itay Bentov
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
222
|
Abstract
A few signaling pathways are driving the growth of hepatocellular carcinoma. Each of these pathways possesses negative regulators. These enzymes, which normally suppress unchecked cell proliferation, are circumvented in the oncogenic process, either the over-activity of oncogenes is sufficient to annihilate the activity of tumor suppressors or tumor suppressors have been rendered ineffective. The loss of several key tumor suppressors has been described in hepatocellular carcinoma. Here, we systematically review the evidence implicating tumor suppressors in the development of hepatocellular carcinoma.
Collapse
|
223
|
DiFeo A, Feld L, Rodriguez E, Wang C, Beer DG, Martignetti JA, Narla G. A functional role for KLF6-SV1 in lung adenocarcinoma prognosis and chemotherapy response. Cancer Res 2008; 68:965-70. [PMID: 18250346 PMCID: PMC2826216 DOI: 10.1158/0008-5472.can-07-2604] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kruppel-like factor 6 (KLF6) is a tumor suppressor gene that is functionally inactivated in human cancer by loss of heterozygosity, somatic mutation, decreased expression, and increased alternative splicing into an oncogenic splice variant, KLF6-SV1. Here we show that increased expression of KLF6-SV1 is associated with decreased survival in patients with lung adenocarcinoma. In addition, KLF6-SV1 is a novel antiapoptotic protein in lung cancer cell lines, and targeted reduction of KLF6-SV1 using siRNA induces apoptosis both alone and in combination with the chemotherapeutic drug cisplatin. Together, these findings highlight a critical role for KLF6-SV1 in lung cancer, and show a potential novel therapeutic strategy for the treatment of lung cancer.
Collapse
Affiliation(s)
- Analisa DiFeo
- Department of Genetics and Genomic Sciences, New York, New York
| | - Lauren Feld
- Department of Genetics and Genomic Sciences, New York, New York
| | | | - Christine Wang
- Department of Genetics and Genomic Sciences, New York, New York
| | - David G. Beer
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - John A. Martignetti
- Department of Genetics and Genomic Sciences, New York, New York
- Department of Oncological Sciences, New York, New York
| | - Goutham Narla
- Department of Genetics and Genomic Sciences, New York, New York
- Department of Medicine, the Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
224
|
Xu J, Lü B, Xu F, Gu H, Fang Y, Huang Q, Lai M. Dynamic down-regulation of Krüppel-like factor 4 in colorectal adenoma-carcinoma sequence. J Cancer Res Clin Oncol 2008; 134:891-8. [PMID: 18264726 DOI: 10.1007/s00432-008-0353-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 01/08/2008] [Indexed: 01/06/2023]
Abstract
PURPOSE To explore the clinical significance of Krüppel-like factors 4 (KLF4) expression in colorectal cancer initiation and progression. METHODS We used quantitative real-time PCR to detect KLF4 mRNA expression in 49 colorectal cancer samples with individual-matched normal mucosa and eight concurrent adenomas. We also analysed the immunostaining pattern of KLF4 in additional 129 colorectal cancers and 48 sporadic colorectal adenomas with matched normal mucosa and correlated KLF4 staining with clinicopathological parameters and prognosis. KLF4 expression change was detected in SW480, SW620 and RKO cell lines after treatment of 5-aza-dC (10 microM) or butyrate sodium (4 mM). RESULTS The large clinicopathological survey with combined methods confirmed a dynamic downregulation of KLF4 in individual-matched normal mucosa, adenoma and cancer (P < 0.05). The quantitative analysis of immunostaining pattern showed that KLF4 staining cells were more frequently seen in the upper zones than that in the lower zones of both normal mucosa and adenoma (P < 0.05). Survival analysis implied a trend toward better overall survival in KLF4-positive colorectal cancer patients with lymph node metastasis than that in KLF4-negative cancer with lymph node metastasis. In vitro study found elevated KLF4 mRNA expression in SW620 and RKO cells with 5-aza-dC treatment, implicating the underlying aberrant epigenetic modifications in regulating KLF4 expression at least in a subset of colorectal cancers. CONCLUSIONS KLF4 is associated with terminal differentiation in colorectal epithelium and drastically downregulated in colorectal adenomas and cancers via possible epigenetic modifications. Loss of KLF4 protein expression might contribute to assessing prognosis in colorectal cancer with lymph node metastasis.
Collapse
Affiliation(s)
- Jing Xu
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
225
|
Lin WM, Baker AC, Beroukhim R, Winckler W, Feng W, Marmion JM, Laine E, Greulich H, Tseng H, Gates C, Hodi FS, Dranoff G, Sellers WR, Thomas RK, Meyerson M, Golub TR, Dummer R, Herlyn M, Getz G, Garraway LA. Modeling genomic diversity and tumor dependency in malignant melanoma. Cancer Res 2008; 68:664-73. [PMID: 18245465 PMCID: PMC10493008 DOI: 10.1158/0008-5472.can-07-2615] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The classification of human tumors based on molecular criteria offers tremendous clinical potential; however, discerning critical and "druggable" effectors on a large scale will also require robust experimental models reflective of tumor genomic diversity. Here, we describe a comprehensive genomic analysis of 101 melanoma short-term cultures and cell lines. Using an analytic approach designed to enrich for putative "driver" events, we show that cultured melanoma cells encompass the spectrum of significant genomic alterations present in primary tumors. When annotated according to these lesions, melanomas cluster into subgroups suggestive of distinct oncogenic mechanisms. Integrating gene expression data suggests novel candidate effector genes linked to recurrent copy gains and losses, including both phosphatase and tensin homologue (PTEN)-dependent and PTEN-independent tumor suppressor mechanisms associated with chromosome 10 deletions. Finally, sample-matched pharmacologic data show that FGFR1 mutations and extracellular signal-regulated kinase (ERK) activation may modulate sensitivity to mitogen-activated protein kinase/ERK kinase inhibitors. Genetically defined cell culture collections therefore offer a rich framework for systematic functional studies in melanoma and other tumors.
Collapse
Affiliation(s)
- William M. Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Alissa C. Baker
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Wendy Winckler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Whei Feng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | | | - Elisabeth Laine
- Department of Dermatology, University of Zurich Hospital, Zürich, Switzerland
| | - Heidi Greulich
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Hsiuyi Tseng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
| | | | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | - William R. Sellers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Roman K. Thomas
- Max Planck Institute for Neurological Research with Klaus Joachim Zulch Laboratories of the Max Planck Society and the Medical Faculty of the University of Cologne
- Center for Integrated Oncology and Department I for Internal Medicine, University of Cologne, Cologne, Germany
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
- The Broad Institute of M.I.T. and Harvard
| | - Todd R. Golub
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Reinhard Dummer
- Department of Dermatology, University of Zurich Hospital, Zürich, Switzerland
| | - Meenhard Herlyn
- Cancer Biology Division, Wistar Institute, Philadelphia, Pennsylvania
| | - Gad Getz
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| | - Levi A. Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Department of Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Harvard Medical School
- The Broad Institute of M.I.T. and Harvard
| |
Collapse
|
226
|
Solé X, Hernández P, de Heredia ML, Armengol L, Rodríguez-Santiago B, Gómez L, Maxwell CA, Aguiló F, Condom E, Abril J, Pérez-Jurado L, Estivill X, Nunes V, Capellá G, Gruber SB, Moreno V, Pujana MA. Genetic and genomic analysis modeling of germline c-MYC overexpression and cancer susceptibility. BMC Genomics 2008; 9:12. [PMID: 18190704 PMCID: PMC2244606 DOI: 10.1186/1471-2164-9-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 01/11/2008] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Germline genetic variation is associated with the differential expression of many human genes. The phenotypic effects of this type of variation may be important when considering susceptibility to common genetic diseases. Three regions at 8q24 have recently been identified to independently confer risk of prostate cancer. Variation at 8q24 has also recently been associated with risk of breast and colorectal cancer. However, none of the risk variants map at or relatively close to known genes, with c-MYC mapping a few hundred kilobases distally. RESULTS This study identifies cis-regulators of germline c-MYC expression in immortalized lymphocytes of HapMap individuals. Quantitative analysis of c-MYC expression in normal prostate tissues suggests an association between overexpression and variants in Region 1 of prostate cancer risk. Somatic c-MYC overexpression correlates with prostate cancer progression and more aggressive tumor forms, which was also a pathological variable associated with Region 1. Expression profiling analysis and modeling of transcriptional regulatory networks predicts a functional association between MYC and the prostate tumor suppressor KLF6. Analysis of MYC/Myc-driven cell transformation and tumorigenesis substantiates a model in which MYC overexpression promotes transformation by down-regulating KLF6. In this model, a feedback loop through E-cadherin down-regulation causes further transactivation of c-MYC. CONCLUSION This study proposes that variation at putative 8q24 cis-regulator(s) of transcription can significantly alter germline c-MYC expression levels and, thus, contribute to prostate cancer susceptibility by down-regulating the prostate tumor suppressor KLF6 gene.
Collapse
Affiliation(s)
- Xavier Solé
- Bioinformatics and Biostatistics Unit, and Translational Research Laboratory, Catalan Institute of Oncology, IDIBELL, L'Hospitalet, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Silencing of the transcription factor KLF6 by siRNA leads to cell cycle arrest and sensitizes cells to apoptosis induced by DNA damage. Cell Death Differ 2008; 15:613-6. [DOI: 10.1038/sj.cdd.4402299] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
228
|
Bureau C, Péron JM, Bouisson M, Danjoux M, Selves J, Bioulac-Sage P, Balabaud C, Torrisani J, Cordelier P, Buscail L, Vinel JP. Expression of the transcription factor Klf6 in cirrhosis, macronodules, and hepatocellular carcinoma. J Gastroenterol Hepatol 2008; 23:78-86. [PMID: 18171345 DOI: 10.1111/j.1440-1746.2007.05234.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Macronodules (MN) occurring in cirrhosis are considered to be precursor lesions for hepatocellular carcinoma (HCC). However, early molecular events in hepatocellular carcinogenesis are poorly understood. The aim of this study was to compare gene expression profiling between cirrhotic tissues, MN, and HCC, to identify genes early involved in liver carcinogenesis. METHODS Tissues were obtained from explanted livers: nine cirrhosis, 10 MN, and seven HCC. Total RNAs were extracted by RNeasy and reverse transcribed with labelled [(33)P]-alpha ATP. Hybridations were performed on Atlas Human Cancer 1.2 membranes (1176 genes). RESULTS A two-way hierarchical clustering algorithm successfully isolated specific gene expression profiles when comparing MN, cirrhosis, and HCC. A total of 16 and 14 genes were up- and down-expressed, respectively, in HCC as compared to cirrhotic tissues. The molecular signature of MN was characterized by the down-expression of 23 and 42 genes as compared to cirrhosis and HCC, respectively. Among them, Klf6 was down-expressed in all MN samples whereas it was over-expressed in cirrhosis and HCC. This result was confirmed at RNA level by quantitative real time-polymerase chain reaction and at protein level by Western blotting. However, no mutation in the exon 2 of Klf6 was detected. CONCLUSION We identified a molecular signature of MN characterized by a down-expression of several genes. One of them, Klf6 was found to be down-expressed in all MN without evidence of somatic mutations in the exon 2. This gene could be involved at an early stage of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Christophe Bureau
- INSERM U858, Fédération Digestive Purpan, CHU Toulouse, Toulouse, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Microarrays--identifying molecular portraits for prostate tumors with different Gleason patterns. METHODS IN MOLECULAR MEDICINE 2008; 141:131-51. [PMID: 18453088 DOI: 10.1007/978-1-60327-148-6_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We present in this chapter the combined use of several recently introduced methodologies for the analysis of microarray datasets. These computational techniques are varied in type and very powerful when combined. We have selected a prostate cancer dataset which is available in the public domain to allow for further comparisons with existing methods. The task is to identify biomarkers that correlate with the clinical phenotype of interest, i.e., Gleason patterns 3, 4, and 5. A supervised method, based on the mathematical formalism of (alpha, beta)-k-feature sets (1), is used to select differentially expressed genes. After these "molecular signatures" are identified, we applied an unsupervised method (a memetic algorithm) to order the samples (2). The objective is to maximize a global measure of correlation in the two-dimensional display of gene expression profiles. With the resulting ordering and taxonomy we are able to identify samples that have been assigned a certain Gleason pattern, and have gene expression patterns different from most of the other samples in the group. We reiterate the approach to obtain molecular signatures that produce coherent patterns of gene expression in each of the three Gleason pattern groups, and we analyze the statistically significant patterns of gene expression that seem to be implicated in these different stages of disease.
Collapse
|
230
|
Abstract
The hepatic stellate cell has surprised and engaged physiologists, pathologists, and hepatologists for over 130 years, yet clear evidence of its role in hepatic injury and fibrosis only emerged following the refinement of methods for its isolation and characterization. The paradigm in liver injury of activation of quiescent vitamin A-rich stellate cells into proliferative, contractile, and fibrogenic myofibroblasts has launched an era of astonishing progress in understanding the mechanistic basis of hepatic fibrosis progression and regression. But this simple paradigm has now yielded to a remarkably broad appreciation of the cell's functions not only in liver injury, but also in hepatic development, regeneration, xenobiotic responses, intermediary metabolism, and immunoregulation. Among the most exciting prospects is that stellate cells are essential for hepatic progenitor cell amplification and differentiation. Equally intriguing is the remarkable plasticity of stellate cells, not only in their variable intermediate filament phenotype, but also in their functions. Stellate cells can be viewed as the nexus in a complex sinusoidal milieu that requires tightly regulated autocrine and paracrine cross-talk, rapid responses to evolving extracellular matrix content, and exquisite responsiveness to the metabolic needs imposed by liver growth and repair. Moreover, roles vital to systemic homeostasis include their storage and mobilization of retinoids, their emerging capacity for antigen presentation and induction of tolerance, as well as their emerging relationship to bone marrow-derived cells. As interest in this cell type intensifies, more surprises and mysteries are sure to unfold that will ultimately benefit our understanding of liver physiology and the diagnosis and treatment of liver disease.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York 10029-6574, USA.
| |
Collapse
|
231
|
Prostate Molecular Oncogenesis. Prostate Cancer 2008. [DOI: 10.1007/978-1-60327-079-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
232
|
Wong CCL, Ng IOL. Gene expression profiles of different stages of hepatocarcinogenesis. J Gastroenterol Hepatol 2008; 23:1-3. [PMID: 18171335 DOI: 10.1111/j.1440-1746.2007.05211.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
233
|
Cheng XF, Li D, Zhuang M, Chen ZY, Lu DX, Hattori T. Growth Inhibitory Effect of Krueppel-like Factor 6 on Human Prostatic Carcinoma and Renal Carcinoma Cell Lines. TOHOKU J EXP MED 2008; 216:35-45. [DOI: 10.1620/tjem.216.35] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Xian-Feng Cheng
- Department of Urology, Second Affiliated Hospital of Harbin Medical University
- Department of Urology, Heilongjiang Province Hospital
| | - Di Li
- Department of Microbiology, Harbin Medical University
| | - Min Zhuang
- Department of Microbiology, Harbin Medical University
| | - Zhao-Yan Chen
- Department of Urology, Second Affiliated Hospital of Harbin Medical University
| | - De-Xiang Lu
- Department of Urology, Heilongjiang Province Hospital
| | - Toshio Hattori
- Division of Emerging Infectious Disease, Tohoku University School of Medicine
| |
Collapse
|
234
|
Guo H, Lin Y, Zhang H, Liu J, Zhang N, Li Y, Kong D, Tang Q, Ma D. Tissue factor pathway inhibitor-2 was repressed by CpG hypermethylation through inhibition of KLF6 binding in highly invasive breast cancer cells. BMC Mol Biol 2007; 8:110. [PMID: 18053161 PMCID: PMC2233638 DOI: 10.1186/1471-2199-8-110] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 12/03/2007] [Indexed: 12/16/2022] Open
Abstract
Background Tissue factor pathway inhibitor-2 (TFPI-2) is a matrix-associated Kunitz inhibitor that inhibits plasmin and trypsin-mediated activation of zymogen matrix metalloproteinases involved in tumor progression, invasion and metastasis. Here, we have investigated the mechanism of DNA methylation on the repression of TFPI-2 in breast cancer cell lines. Results We found that both protein and mRNA of TFPI-2 could not be detected in highly invasive breast cancer cell line MDA-MB-435. To further investigate the mechanism of TFPI-2 repression in breast cancer cells, 1.5 Kb TFPI-2 promoter was cloned, and several genetic variations were detected, but the promoter luciferase activities were not affected by the point mutation in the promoter region and the phenomena was further supported by deleted mutation. Scan mutation and informatics analysis identified a potential KLF6 binding site in TFPI-2 promoter. It was revealed, by bisulfite modified sequence, that the CpG island in TFPI-2 promoter region was hypermethylated in MDA-MB-435. Finally, using EMSA and ChIP assay, we demonstrated that the CpG methylation in the binding site of KLF-6 diminished the binding of KLF6 to TFPI-2 promoter. Conclusion In this study, we found that the CpG islands in TFPI-2 promoter was hypermethylated in highly invasive breast cancer cell line, and DNA methylation in the entire promoter region caused TFPI-2 repression by inducing inactive chromatin structure and decreasing KLF6 binding to its DNA binding sequence.
Collapse
Affiliation(s)
- Hongshen Guo
- Key Laboratory of Molecular Medicine, Ministry of Education, Yixueyuan Road 138#, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Wong OGW, Nitkunan T, Oinuma I, Zhou C, Blanc V, Brown RSD, Bott SRJ, Nariculam J, Box G, Munson P, Constantinou J, Feneley MR, Klocker H, Eccles SA, Negishi M, Freeman A, Masters JR, Williamson M. Plexin-B1 mutations in prostate cancer. Proc Natl Acad Sci U S A 2007; 104:19040-5. [PMID: 18024597 PMCID: PMC2141904 DOI: 10.1073/pnas.0702544104] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Indexed: 11/18/2022] Open
Abstract
Semaphorins are a large class of secreted or membrane-associated proteins that act as chemotactic cues for cell movement via their transmembrane receptors, plexins. We hypothesized that the function of the semaphorin signaling pathway in the control of cell migration could be harnessed by cancer cells during invasion and metastasis. We now report 13 somatic missense mutations in the cytoplasmic domain of the Plexin-B1 gene. Mutations were found in 89% (8 of 9) of prostate cancer bone metastases, in 41% (7 of 17) of lymph node metastases, and in 46% (41 of 89) of primary cancers. Forty percent of prostate cancers contained the same mutation. Overexpression of the Plexin-B1 protein was found in the majority of primary tumors. The mutations hinder Rac and R-Ras binding and R-RasGAP activity, resulting in an increase in cell motility, invasion, adhesion, and lamellipodia extension. These results identify a key role for Plexin-B1 and the semaphorin signaling pathway it mediates in prostate cancer.
Collapse
Affiliation(s)
- Oscar Gee-Wan Wong
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Tharani Nitkunan
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Izumi Oinuma
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Chun Zhou
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Veronique Blanc
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Richard S. D. Brown
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Simon R. J. Bott
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Joseph Nariculam
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Gary Box
- Institute of Cancer Research, Cotswold Road, Sutton, Surrey SM2 5NG, United Kingdom
| | - Phillipa Munson
- Department of Histopathology, University College London, London WC1E 6BT, United Kingdom; and
| | - Jason Constantinou
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Mark R. Feneley
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Helmut Klocker
- Department of Urology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Suzanne A. Eccles
- Institute of Cancer Research, Cotswold Road, Sutton, Surrey SM2 5NG, United Kingdom
| | - Manabu Negishi
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Alex Freeman
- Department of Histopathology, University College London, London WC1E 6BT, United Kingdom; and
| | - John R. Masters
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| | - Magali Williamson
- *Prostate Cancer Research Centre, Institute of Urology, University College London, 67 Riding House Street, London W1W 7EJ, United Kingdom
| |
Collapse
|
236
|
Caffeine regulates alternative splicing in a subset of cancer-associated genes: a role for SC35. Mol Cell Biol 2007; 28:883-95. [PMID: 18025108 DOI: 10.1128/mcb.01345-07] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alternative splicing of pre-mRNA contributes significantly to human proteomic complexity, playing a key role in development, gene expression and, when aberrant, human disease onset. Many of the factors involved in alternative splicing have been identified, but little is known about their regulation. Here we report that caffeine regulates alternative splicing of a subset of cancer-associated genes, including the tumor suppressor KLF6. This regulation is at the level of splice site selection, occurs rapidly and reversibly, and is concentration dependent. We have recapitulated caffeine-induced alternative splicing of KLF6 using a cell-based minigene assay and identified a "caffeine response element" within the KLF6 intronic sequence. Significantly, a chimeric minigene splicing assay demonstrated that this caffeine response element is functional in a heterologous context; similar elements exist within close proximity to caffeine-regulated exons of other genes in the subset. Furthermore, the SR splicing factor, SC35, was shown to be required for induction of the alternatively spliced KLF6 transcript. Importantly, SC35 is markedly induced by caffeine, and overexpression of SC35 is sufficient to mimic the effect of caffeine on KLF6 alternative splicing. Taken together, our data implicate SC35 as a key player in caffeine-mediated splicing regulation. This novel effect of caffeine provides a valuable tool for dissecting the regulation of alternative splicing of a large gene subset and may have implications with respect to splice variants associated with disease states.
Collapse
|
237
|
Glinsky GV. Stem cell origin of death-from-cancer phenotypes of human prostate and breast cancers. ACTA ACUST UNITED AC 2007; 3:79-93. [PMID: 17873385 DOI: 10.1007/s12015-007-0011-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/01/2022]
Abstract
In clinical terms, all human cancers diagnosed in individuals can be divided in two major categories: malignant tumors that will be cured with the existing cancer therapies and tumors that have therapy-resistant phenotypes and will return after initial treatment as incurable metastatic disease. These tumors manifesting clinically lethal death-from-cancer phenotypes represent the most formidable challenge of experimental, translational, and clinical cancer research. Clinical genomics data demonstrate that gene expression signatures associated with the "stemness" state of a cell are informative as molecular predictors of cancer therapy outcome and can help to identify cancer patients with therapy-resistant tumors. Here, we present experimental and clinical evidence in support of the BMI1 pathway rule indicating a genetic link between the stemness state and therapy-resistant death-from-cancer phenotypes. Our analysis demonstrates that therapy-resistant and therapy-responsive cancer phenotypes manifest distinct patterns of association with stemness/differentiation pathways, suggesting that therapy-resistant and therapy-responsive tumors develop within genetically distinct stemness/differentiation programs. These differences can be exploited for development of prognostic and therapy selection genetic tests utilizing a microarray-based cancer therapy outcome predictor algorithm. One of the major regulatory pathways manifesting distinct patterns of association with therapy-resistant and therapy-responsive cancer phenotypes is the Polycomb group proteins chromatin silencing pathway.
Collapse
Affiliation(s)
- Gennadi V Glinsky
- Translational & Functional Genomics Laboratory, Ordway Cancer Center, Ordway Research Institute, Albany, NY 12208, USA.
| |
Collapse
|
238
|
Veldman MB, Bemben MA, Thompson RC, Goldman D. Gene expression analysis of zebrafish retinal ganglion cells during optic nerve regeneration identifies KLF6a and KLF7a as important regulators of axon regeneration. Dev Biol 2007; 312:596-612. [PMID: 17949705 DOI: 10.1016/j.ydbio.2007.09.019] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 09/11/2007] [Accepted: 09/13/2007] [Indexed: 01/07/2023]
Abstract
Unlike mammals, teleost fish are able to mount an efficient and robust regenerative response following optic nerve injury. Although it is clear that changes in gene expression accompany axonal regeneration, the extent of this genomic response is not known. To identify genes involved in successful nerve regeneration, we analyzed gene expression in zebrafish retinal ganglion cells (RGCs) regenerating their axons following optic nerve injury. Microarray analysis of RNA isolated by laser capture microdissection from uninjured and 3-day post-optic nerve injured RGCs identified 347 up-regulated and 29 down-regulated genes. Quantitative RT-PCR and in situ hybridization were used to verify the change in expression of 19 genes in this set. Gene ontological analysis of the data set suggests regenerating neurons up-regulate genes associated with RGC development. However, not all regeneration-associated genes are expressed in differentiating RGCs indicating the regeneration is not simply a recapitulation of development. Knockdown of six highly induced regeneration-associated genes identified two, KLF6a and KLF7a, that together were necessary for robust RGC axon re-growth. These results implicate KLF6a and KLF7a as important mediators of optic nerve regeneration and suggest that not all induced genes are essential to mount a regenerative response.
Collapse
Affiliation(s)
- Matthew B Veldman
- Neuroscience Program, University of Michigan, 5045 Biomedical Sciences Research Building, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
239
|
Filion E, Taussky D, Bahary JP, Maugard CM. Higher frequency of familial clustering of prostate cancer in French-Canadian men. J Urol 2007; 178:1265-9; discussion 1270. [PMID: 17698103 DOI: 10.1016/j.juro.2007.05.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Indexed: 11/22/2022]
Abstract
PURPOSE Prostate cancer is the second cause of cancer related death in North American men. We investigated the frequency of familial clustering in a French-Canadian population of prostate cancer cases. MATERIALS AND METHODS Between October 2004 and September 2005, 179 consecutively seen patients with localized prostate cancer identified each of their parents as being of French-Canadian descent. They were asked for their family history of cancer in first-degree relatives, age at diagnosis, whether affected relatives were alive, age and markers of tumor aggressiveness, including prostate specific antigen, Gleason and disease stage. ANOVA was used to compare the distribution of quantitative factors according to qualitative factors identified in our population. Differences between qualitative factors were assessed by the Fisher exact test. All p values were 2-sided. RESULTS Mean age at diagnosis was 67 years. A total of 45 French-Canadian patients (25.1%) had at least 1 first-degree relative with prostate cancer, including 34 (19%) with 1 first-degree relative, 9 with a father-son pair, 25 with a brother-brother pair and 11 (6.1%) with at least 2 first-degree relatives. In our series the frequency of familial clustering defined by at least 1 relative with prostate cancer was high. We found a higher percent of French-Canadian men with at least 1 first-degree relative with prostate cancer than what was previously reported for an unselected population in Canada (25.1% vs 14.7%, p <0.0001). CONCLUSIONS Those preliminary results open a new perspective to a better understanding of familial prostate cancer in the Province of Quebec.
Collapse
Affiliation(s)
- Edith Filion
- Département de Radio-Oncologie, Centre Hospitalier Université de Montréal, Montréal, Québec, Canada.
| | | | | | | |
Collapse
|
240
|
Mukai S, Hiyama T, Tanaka S, Yoshihara M, Arihiro K, Chayama K. Involvement of Krüppel-like factor 6 ( KLF6) mutation in the development of nonpolypoid colorectal carcinoma. World J Gastroenterol 2007; 13:3932-8. [PMID: 17663506 PMCID: PMC4171164 DOI: 10.3748/wjg.v13.i29.3932] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine Krüppel-like factor 6 (KLF6) mutations in nonpolypoid-type tumors and alterations of K-ras, p53, and B-raf in relation between mutation and morphologic type, particularly nonpolypoid-type colorectal carcinomas.
METHODS: Fifty-five early nonpolypoid colorectal carcinomas were analyzed. Loss of heterozygosity (LOH) of KLF6 and p53 was determined by microsatellite assay. Mutations of KLF6, K-ras, and B-raf were examined by polymerase chain reaction-single-strand conformation polymorphism followed by direct sequencing. In LOH-positive and/or mutation-positive tumors, multiple (4-7) samples in each tumor were microdissected and examined for genetic alterations. p53 expression was evaluated by immunohistochemistry.
RESULTS: LOH of KLF6 and p53 was found in 14 of 29 (48.3%) and 14 of 31 (45.2%) tumors, respectively. In 10 of the 14 (71.4%) KLF6 LOH-positive tumors and 9 of the 14 (64.3%) p53 LOH-positive tumors, LOH was found in all of the microdissected samples. In 1 of the 10 (10.0%) KLF6 LOH-positive tumors, a single missense mutation was identified. K-ras and B-raf mutations were found in 5 of 55 (9.1%) and 6 of 55 (10.9%) tumors, respectively. However, these mutations were detected only in subsets of microdissected tumor samples.
CONCLUSION: These data suggest that KLF6 and p53 mutations are involved in the development of nonpolypoid colorectal carcinoma, whereas K-ras and B-raf mutations are not.
Collapse
Affiliation(s)
- Shinichi Mukai
- Department of Endoscopy, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | | | | | | | | | | |
Collapse
|
241
|
Chen C, Benjamin MS, Sun X, Otto KB, Guo P, Dong XY, Bao Y, Zhou Z, Cheng X, Simons JW, Dong JT. KLF5 promotes cell proliferation and tumorigenesis through gene regulation and the TSU-Pr1 human bladder cancer cell line. Int J Cancer 2007; 118:1346-55. [PMID: 16184550 DOI: 10.1002/ijc.21533] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
KLF5 is a transcription factor that plays important roles in multiple physical and pathological processes, including cell growth, cell cycle regulation, and angiogenesis. To better characterize KLF5 function in bladder carcinogenesis, we established stable TSU-Pr1 cell clones expressing different levels of KLF5. These clones were then characterized for cell growth, cell cycle progression, tumorigenesis, and alteration in gene expression. Overexpression of KLF5 promoted tumorigenesis of the TSU-Pr1 cancer cells in mice. Consistently, KLF5 increased G1 to S phase transition, which was accompanied by the upregulation of cyclin D1, phosphorylation of MAPK and Akt, and reduced protein levels for CDK inhibitors p27 and p15. Microarray analysis combined with expression verification in different cell systems identified a number of additional genes that are potentially regulated by KLF5, including HBP17, ITGA6, and RAIG1. These findings suggest that the KLF5 transcription factor plays an oncogenic role in the TSU-Pr1 bladder cancer cell line through the regulation of a subset of genes.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation
- Clone Cells
- Cyclin D1/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/physiology
- Male
- Mice
- Mice, SCID
- Mitogen-Activated Protein Kinases/metabolism
- Neoplasm Transplantation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Oligonucleotide Array Sequence Analysis
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- S Phase/genetics
- S Phase/physiology
- Transplantation, Heterologous
- Tumor Burden
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Ceshi Chen
- Winship Cancer Institute and Department of Oncology and Hematology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
O'Grady E, Mulcahy H, Adams C, Morrissey JP, O'Gara F. Manipulation of host Kruppel-like factor (KLF) function by exotoxins from diverse bacterial pathogens. Nat Rev Microbiol 2007; 5:337-41. [PMID: 17435789 DOI: 10.1038/nrmicro1641] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diverse pathogenic bacteria have developed similar mechanisms to subvert host cell responses. In this Progress article, we focus on bacterial virulence factors with different enzymatic activities that can increase the expression of the Kruppel-like factor (KLF) family of mammalian transcriptional regulators through their ability to modify the activity of a common host-cell target - the Rho protein family. By using a common virulence strategy, both Gram-negative and Gram-positive pathogens exploit the KLF regulatory cascade to modulate nuclear factor kappaB activation, pro-inflammatory cytokine expression, actin cytoskeletal dynamics and phagocytosis.
Collapse
Affiliation(s)
- Eoin O'Grady
- BIOMERIT Research Centre, Department of Microbiology, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
243
|
Aitchison A, Warren A, Neal D, Rabbitts P. RASSF1A promoter methylation is frequently detected in both pre-malignant and non-malignant microdissected prostatic epithelial tissues. Prostate 2007; 67:638-44. [PMID: 17342751 DOI: 10.1002/pros.20475] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The RASSF1A gene is a tumor suppressor gene inactivated by hypermethylation in a very wide variety of malignant tumors including prostate cancer. METHODS In this study we have used laser capture microdissection to provide pure cell populations to investigate the methylation status of 16 CpG sites in the promoter region of this gene in prostatic intra-epithelial neoplasia, in histologically normal epithelial cells associated with these lesions and in epithelial cells from benign prostatic hyperplasia. RESULTS Unexpectedly, frequent methylation, detected by sequence analysis following bisulphite treatment, was observed in benign epithelium as well as in the lesions associated with prostatic intra-epithelial neoplasia and at high risk of cancer formation. Fifty percent or more CpG sites were methylated in 7/14 prostatic intra-epithelial neoplasms, 8/11 histologically normal epithelial cells and 8/12 specimens of benign prostatic tissue. CONCLUSION These observations suggest that methylation of the RASSF1A gene is present in both pre-malignant and benign epithelia and suggests quantitation is required for it to be an effective marker of early prostate cancer.
Collapse
Affiliation(s)
- Alan Aitchison
- Department of Oncology, University of Cambridge, Hills Road, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
244
|
Miyaki M, Yamaguchi T, Iijima T, Funata N, Mori T. Difference in the Role of Loss of Heterozygosity at 10p15 (KLF6 Locus) in Colorectal Carcinogenesis between Sporadic and Familial Adenomatous Polyposis and Hereditary Nonpolyposis Colorectal Cancer Patients. Oncology 2007; 71:131-5. [PMID: 17347589 DOI: 10.1159/000100523] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 11/18/2006] [Indexed: 01/16/2023]
Abstract
OBJECTIVES To clarify the role of the KLF6 (Kruppel-like factor 6) locus in multistep colorectal carcinogenesis, we analyzed loss of heterozygosity (LOH) at 10p15 (KLF6 locus) and mutations of the KLF6gene in 298 colorectal tumors at various pathological stages of sporadic and familial adenomatous polyposis (FAP) and hereditary nonpolyposis colorectal cancer (HNPCC) patients. METHODS 10p15 LOH was analyzed using KLF6M1 and KLF6M2, and KLF6 gene mutation was analyzed using PCR-SSCP and sequencing. RESULTS It was found that the frequencies of LOH (sum of M1 and M2) were 4% in adenomas, 0% in intramucosal carcinomas, 35% in invasive carcinomas, and 33% in liver metastases in sporadic cases. Invasive carcinomas from FAP patients showed only 6% LOH, and invasive carcinomas from HNPCC patients exhibited 0% LOH. Mutation analysis of the KLF6 gene in 298 colorectal tumors detected no somatic mutations. CONCLUSIONS The present data suggest that LOH of the KLF6 locus at chromosome 10p15 contributes to the invasion step from an intramucosal carcinoma to an invasive carcinoma specifically in sporadic colorectal carcinogenesis, but is rarely involved in the carcinogenesis of FAP and HNPCC cases. Moreover, the absence of somatic mutations suggests the uncertainty of the KLF6 gene as a classical tumor suppressor gene at the lost 10p15 region in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Michiko Miyaki
- Hereditary Tumor Research Project, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | | | | | | | | |
Collapse
|
245
|
|
246
|
Kremer-Tal S, Narla G, Chen Y, Hod E, DiFeo A, Yea S, Lee JS, Schwartz M, Thung SN, Fiel IM, Banck M, Zimran E, Thorgeirsson SS, Mazzaferro V, Bruix J, Martignetti JA, Llovet JM, Friedman SL. Downregulation of KLF6 is an early event in hepatocarcinogenesis, and stimulates proliferation while reducing differentiation. J Hepatol 2007; 46:645-54. [PMID: 17196295 PMCID: PMC3533246 DOI: 10.1016/j.jhep.2006.10.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 09/19/2006] [Accepted: 10/09/2006] [Indexed: 01/11/2023]
Abstract
BACKGROUND/AIMS Hepatocellular carcinoma (HCC) has the most rapidly rising cancer incidence in the US and Europe. The KLF6 tumor suppressor is frequently inactivated in HCC by loss-of-heterozygosity (LOH) and/or mutation. METHODS Here we have analyzed 33 HBV- and 40 HCV-related HCCs for mRNA expression of wildtype KLF6 (wtKLF6) as well as the KLF6 variant 1 (SV1), a truncated, growth-promoting variant that antagonizes wtKLF6 function. The HCV-related tumors analyzed represented the full histologic spectrum from cirrhosis and dysplasia to metastatic cancer. RESULTS Expression of KLF6 mRNA is decreased in 73% of HBV-associated HCCs compared to matched surrounding tissue (ST), with reductions of approximately 80% in one-third of the patients. KLF6 mRNA expression is also reduced in dysplastic nodules from patients with HCV compared to cirrhotic livers (p<0.005), with an additional, marked decrease in the very advanced, metastatic stage (p<0.05). An increased ratio of KLF6SV1/wt KLF6 is present in a subset (6/33, 18%) of the HBV-related HCCs compared to matched ST. Reconstituting KLF6 in HepG2 cells by retroviral infection decreased proliferation and related markers including cyclin D1 and beta-catenin, increased cellular differentiation based on induction of albumin, E-cadherin, and decreased alpha fetoprotein. CONCLUSIONS We conclude that reduced KLF6 expression is common in both HBV- and HCV-related HCCs and occurs at critical stages during cancer progression. Effects of KLF6 are attributable to regulation of genes controlling hepatocyte growth and differentiation.
Collapse
Affiliation(s)
- Sigal Kremer-Tal
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Goutham Narla
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Yingbei Chen
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Eldad Hod
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Analisa DiFeo
- Department of Human Genetics, Mount Sinai School of Medicine, New York, NY, USA
| | - Steven Yea
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Ju-Seog Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Molecular Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Myron Schwartz
- Department of Surgery, Mount Sinai School of Medicine, New York, NY, USA
- Recanati-Miller Transplantation Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Swan N. Thung
- Department of Pathology, Mount Sinai School of Medicine, New York, NY, USA
| | - Isabel M. Fiel
- Department of Pathology, Mount Sinai School of Medicine, New York, NY, USA
| | - Michaela Banck
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Eran Zimran
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
- Ben-Gurion Medical School, Beer-Sheba, Israel
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Jordi Bruix
- BCLC group, Liver Unit, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - John A. Martignetti
- Department of Human Genetics, Mount Sinai School of Medicine, New York, NY, USA
- Department of Pediatrics, Mount Sinai School of Medicine, New York, NY, USA
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Josep M. Llovet
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
- Recanati-Miller Transplantation Institute, Mount Sinai School of Medicine, New York, NY, USA
- BCLC group, Liver Unit, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Scott L. Friedman
- Division of Liver Diseases and Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
- Corresponding author. Tel.: +1 212 659 9501; fax: +1 212 849 2574., (S.L. Friedman)
| |
Collapse
|
247
|
Sirach E, Bureau C, Péron JM, Pradayrol L, Vinel JP, Buscail L, Cordelier P. KLF6 transcription factor protects hepatocellular carcinoma-derived cells from apoptosis. Cell Death Differ 2007; 14:1202-10. [PMID: 17347668 DOI: 10.1038/sj.cdd.4402114] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major public health concern because of the absence of early diagnosis and effective treatments. Efficient diagnosis modalities and therapies to treat HCC are needed. Kruppel-like factor (KLF) family members, such as KLF6, are involved in cell proliferation and differentiation. KLF6 is inactivated in solid tumors, which may contribute to pathogenesis. However, KLF6 status in HCC is controversial. Thus, we undertook the characterization of KLF6 expression and function in HCC and HCC-derived cell lines. We found that HCC, HepG2 and HuH7 cells expressed KLF6 messenger ribonucleic acid and protein. Next, using RNA interference, we demonstrated that inhibiting KLF6 expression in vitro strongly impaired cell proliferation-induced G1-phase arrest, inhibited cyclin-dependent kinase 4 and cyclin D1 expression, and subsequent retinoblastoma phosphorylation. Finally, KLF6 silencing caused p53 upregulation and inhibited Bcl-xL expression, to induce cell death by apoptosis. Taken together, these data demonstrated that KLF6 is essential for HCC-derived cells to evade apoptosis.
Collapse
Affiliation(s)
- E Sirach
- INSERM U858, I2MR, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
248
|
Suzuki T, Nishi T, Nagino T, Sasaki K, Aizawa K, Kada N, Sawaki D, Munemasa Y, Matsumura T, Muto S, Sata M, Miyagawa K, Horikoshi M, Nagai R. Functional Interaction between the Transcription Factor Krüppel-like Factor 5 and Poly(ADP-ribose) Polymerase-1 in Cardiovascular Apoptosis. J Biol Chem 2007; 282:9895-9901. [PMID: 17283079 DOI: 10.1074/jbc.m608098200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Krüppel-like factor 5 (KLF5) is a transcription factor important in regulation of the cardiovascular response to external stress. KLF5 regulates pathological cell growth, and its acetylation is important for this effect. Its mechanisms of action, however, are still unclear. Analysis in KLF5-deficient mice showed that KLF5 confers apoptotic resistance in vascular lesions. Mechanistic analysis further showed that it specifically interacts with poly(ADP-ribose) polymerase-1 (PARP-1), a nuclear enzyme important in DNA repair and apoptosis. KLF5 interacted with a proteolytic fragment of PARP-1, and acetylation of KLF5 under apoptotic conditions increased their affinity. Moreover, KLF5 wild-type (but not a non-acetylatable point mutant) inhibited apoptosis as induced by the PARP-1 fragment. Collectively, we have found that KLF5 regulates apoptosis and targets PARP-1, and further, for acetylation to regulate these effects. Our findings thus implicate functional interaction between the transcription factor KLF5 and PARP-1 in cardiovascular apoptosis.
Collapse
Affiliation(s)
- Toru Suzuki
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Clinical Bioinformatics, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Toshiya Nishi
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoko Nagino
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kana Sasaki
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kenichi Aizawa
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Nanae Kada
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Daigo Sawaki
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yoshiko Munemasa
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takayoshi Matsumura
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shinsuke Muto
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Laboratory of Developmental Biology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kiyoshi Miyagawa
- Department of Radiation Biology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masami Horikoshi
- Laboratory of Developmental Biology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryozo Nagai
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
249
|
Amit I, Citri A, Shay T, Lu Y, Katz M, Zhang F, Tarcic G, Siwak D, Lahad J, Jacob-Hirsch J, Amariglio N, Vaisman N, Segal E, Rechavi G, Alon U, Mills GB, Domany E, Yarden Y. A module of negative feedback regulators defines growth factor signaling. Nat Genet 2007; 39:503-12. [PMID: 17322878 DOI: 10.1038/ng1987] [Citation(s) in RCA: 361] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 01/24/2007] [Indexed: 11/09/2022]
Abstract
Signaling pathways invoke interplays between forward signaling and feedback to drive robust cellular response. In this study, we address the dynamics of growth factor signaling through profiling of protein phosphorylation and gene expression, demonstrating the presence of a kinetically defined cluster of delayed early genes that function to attenuate the early events of growth factor signaling. Using epidermal growth factor receptor signaling as the major model system and concentrating on regulation of transcription and mRNA stability, we demonstrate that a number of genes within the delayed early gene cluster function as feedback regulators of immediate early genes. Consistent with their role in negative regulation of cell signaling, genes within this cluster are downregulated in diverse tumor types, in correlation with clinical outcome. More generally, our study proposes a mechanistic description of the cellular response to growth factors by defining architectural motifs that underlie the function of signaling networks.
Collapse
Affiliation(s)
- Ido Amit
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Narla G, Kremer-Tal S, Matsumoto N, Zhao X, Yao S, Kelley K, Tarocchi M, Friedman SL. In vivo regulation of p21 by the Kruppel-like factor 6 tumor-suppressor gene in mouse liver and human hepatocellular carcinoma. Oncogene 2007; 26:4428-34. [PMID: 17297474 DOI: 10.1038/sj.onc.1210223] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Kruppel-like factor (KLF) 6 is a tumor-suppressor gene functionally inactivated by loss of heterozygosity, somatic mutation and/or alternative splicing that generates a dominant-negative splice form, KLF6-SV1. Wild-type KLF6 (wtKLF6) expression is decreased in many human malignancies, which correlates with reduced patient survival. Additionally, loss of the KLF6 locus in the absence of somatic mutation in the remaining allele occurs in a number of human cancers, raising the possibility that haploinsufficiency of the KLF6 gene alone contributes to cellular growth dysregulation and tumorigenesis. Our earlier studies identified the cyclin-dependent kinase inhibitor p21 as a transcriptional target of the KLF6 gene in cultured cells, but not in vivo. To address this issue, we have generated two genetic mouse models to define the in vivo role of KLF6 in regulating cell proliferation and p21 expression. Transgenic overexpression of KLF6 in the liver resulted in a runted phenotype with decreased body and liver size, with evidence of decreased hepatocyte proliferation, increased p21 and reduced proliferating cell nuclear antigen expression. In contrast, mice with targeted deletion of one KLF6 allele (KLF6+/-) display increased liver mass with reduced p21 expression, compared to wild type littermates. Moreover, in primary hepatocellular carcinoma samples, there is a significant correlation between wtKLF6 and p21 mRNA expression. Combined, these data suggest that haploinsufficiency of the KLF6 gene may regulate cellular proliferation in vivo through decreased transcriptional activation of the cyclin-dependent kinase inhibitor p21.
Collapse
Affiliation(s)
- G Narla
- Department of Medicine, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|