Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

HMGA1a Recognition Candidate DNA Sequences in Humans

  • Takayuki Manabe ,

    manabe@fujita-hu.ac.jp

    Affiliation Division of Gene Expression Mechanism, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan

  • Taiichi Katayama,

    Affiliation Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, Suita, Osaka, Japan

  • Masaya Tohyama

    Affiliations Department of Anatomy and Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, Suita, Osaka, Japan

Abstract

High mobility group protein A1a (HMGA1a) acts as an architectural transcription factor and influences a diverse array of normal biological processes. It binds AT-rich sequences, and previous reports have demonstrated HMGA1a binding to the authentic promoters of various genes. However, the precise sequences that HMGA1a binds to remain to be clarified. Therefore, in this study, we searched for the sequences with the highest affinity for human HMGA1a using an existing SELEX method, and then compared the identified sequences with known human promoter sequences. Based on our results, we propose the sequences “-(G/A)-G-(A/T)-(A/T)-A-T-T-T-” as HMGA1a-binding candidate sequences. Furthermore, these candidate sequences bound native human HMGA1a from SK-N-SH cells. When candidate sequences were analyzed by performing FASTAs against all known human promoter sequences, 500–900 sequences were hit by each one. Some of the extracted genes have already been proven or suggested as HMGA1a-binding promoters. The candidate sequences presented here represent important information for research into the various roles of HMGA1a, including cell differentiation, death, growth, proliferation, and the pathogenesis of cancer.

Introduction

High mobility group protein A1a (HMGA1a) participates in a wide variety of nuclear processes acting as an architectural transcription factor regulating the expression of numerous genes [1][3]. This protein influences a diverse array of normal biological processes, including cell differentiation, death, growth and proliferation, and is involved in the pathogenesis of cancer via protein–protein and DNA–protein interactions [1][3]. Therefore, HMGA1a protein has been described as the central ‘hub’ of nuclear function [2].

HMGA1a binds AT-rich sequences via its own AT-hook, and functions in a variety of ways [1][3]. Many previous reports have demonstrated HMGA1a binding to the authentic promoters of various genes (for example, human KIT Ligand (hKL) [4], Xeroderma pigmentosum complementation group A [5], Cox2 [6], [7], interferon-β [8], interleukin-10 [9] and -4 [10], iNos/Nos2 [11], c-Fos and SM22α [12]) using DNase I protection assays and/or electrophoretic mobility shift assays (EMSAs). Furthermore, several HMGA1a-regulating genes and pathways have been suggested by microarray analyses [13]. However, although AT-rich sequences exist within authentic gene promoters, their affinity for HMGA1a varies from strong to weak to none at all; even within the same promoter, AT-rich sequences can have vastly differing affinities for HMGA1a [8], [14]. It remains to be clarified exactly which sequences HMGA1a binds to, and whether and how co-factors, structures, and the existence of binding regions on the surface of the DNA-protein complex influence HMGA1a-DNA binding. Therefore, using an existing SELEX method to study all known human promoter sequences, we searched for the sequences with the highest affinity for human HMGA1a.

Results and Discussion

Determination of HMGA1a Recognition Candidate DNA Sequences in Humans

The ratios of the four bases in the synthesized random sequences used in this research, which were placed between T7 sequences, were almost uniform, as a result of a direct sequencing (Figure 1a). When these random sequences of DNA were analyzed using the SELEX method with E. coli.-expressed recombinant HMGA1a [15], the ratio of the four bases became AT-rich, with the frequencies of A and T significantly higher (by about 40%) than the frequencies of G and C (Figure 1b). This result shows that the SELEX system selects specific bases; in the case here, and as reported [1], AT-rich sequences. The relative levels of bases in regions assumed to be recognition sequences was as follows: C<G≪<A/T (Figures S1 and 1c). The bases A and T were twice as common, or more, as the bases C and G (Figures S1 and1c). We propose the sequences “-(G/A)-G-(A/T)-(A/T)-A-T-T-T-” as HMGA1a-binding candidate sequences (Figure 1d). Besides being AT-rich, the inclusion of a GG sequence immediately before the AT-rich sequence is interesting. Indeed, the existence of such a GG sequence in authentic promoters has been reported [3], [8], [10].

thumbnail
Figure 1. HMGA1a Recognition Candidate DNA Sequences by SELEX.

Ratio of bases in synthetic DNA sequences before (A) and after (B) SELEX assays. (C) Ratio of bases in regions of candidate DNA sequences after SELEX assays. (D) Candidate HMGA1a binding sequences are shown.

https://doi.org/10.1371/journal.pone.0008004.g001

The Candidate Sequences Bound Native Human HMGA1a

Native HMGA1a undergoes various post-translational modifications [1], [2]. Therefore, binding of endogenous HMGA1a from human cell nuclear extracts to these candidate sequences was examined by EMSAs (Figures 2A and 2B). Previous reports have demonstrated that HMGA1a expression is significantly increased by hypoxia stimuli in human neuroblastoma SK-N-SH cells, but not in HEK293T or HeLa cells [16], [17]. Using the system described in those reports, binding that was weak under normoxia (Figure 2A, lane 1) became much stronger following hypoxic stimulation (Fig. 2A, lane 2 and Fig. 2B). This increase in binding was prevented by inactivation of HMGA1a in the nuclear extracts using an antibody against it (Figures. 2A and 2B). Therefore, our advocated candidate sequence bound native human HMGA1a from SK-N-SH cells.

thumbnail
Figure 2. Effects of endogenous HMGA1a on binding to the candidate sequences by EMSA.

(A) Radiography of EMSA using nuclear extracts obtained from human neuroblastoma SK-N-SH cells under normoxic (lane 1) or hypoxic (lane 2) conditions, or hypoxia (HMGA1a removal: lane 3). (B) Densitometric quantitative data from (A) shown as the % of the levels in normoxia.

https://doi.org/10.1371/journal.pone.0008004.g002

The Candidate Sequences in All Known Human Promoters by FASTA Analysis

There were eight candidate sequences in total: GGAAATTT, GGATATTT, GGTAATTT, GGTTATTT, AGAAATTT, AGATATTT, AGTAATTT, and AGTTATTT. When all known human promoter sequences were analyzed by performing a FASTA on each sequence, 500–900 sequences were hit by each one (Table S1). It is interesting that two or more candidate sequences were found in many of the extracted gene promoters, while the vast majority of human promoter sequences were not hit by any of the candidate sequences (Table 1). This strongly suggests that these candidate sequences are genuine. Moreover, it is also interesting that some of the genes that have already been proven or suggested to have promoters that bind HMGA1a were extracted (Table 2).

thumbnail
Table 1. Percentage of repetition in other candidate sequences on the hit gene promoters retrieved using each candidate sequence.

https://doi.org/10.1371/journal.pone.0008004.t001

thumbnail
Table 2. List of genes that have already been proven or suggested to have promoters that bind HMGA1a.

https://doi.org/10.1371/journal.pone.0008004.t002

The functions of HMGA1a are diverse and it is known to have a role in disease appearance; thus, the possibility of its becoming a target of treatments has been suggested ([28], Table 3). That is, the candidate sequences proposed by this study may be a blocker of the transcription of cancer-related genes (Table 3), as decoy DNAs. We also reported that a decoy RNA of a specific HMGA1a-binding sequence prevents cell death [29]. In conclusion, the candidate sequences presented here represent important information for research into the various roles of HMGA1a.

thumbnail
Table 3. List of genes that have already been proven to associate with each HMGA1a-related cancer/tumor.

https://doi.org/10.1371/journal.pone.0008004.t003

Materials and Methods

DNA Selection Assay In Vitro (SELEX)

A synthesized DNA (1 pmol) [5′-GGTGATCAGATTCTGATCCA (N31) TGAAGCTTGGATCCGTCGC-3′] molecule containing a 31-nucleotide random sequence (20.7% A, 22.7% C, 31.5% T, 25.1% G by direct sequencing of 16 clones) was amplified (seven cycles) by PCR, followed by incubation with E. coli.-expressed rHMGA1a in incubation buffer [16] for 30 min at 25°C. The reaction solution was then subjected to immunoprecipitation with an antibody against HMGA1, followed by amplification (seven cycles) by PCR. The PCR products were cloned into a pGEM-T vector and analyzed by direct sequencing.

Gel electrophoresis Mobility Shift Assay (EMSA)

After determining the protein content in the nuclear extracts, an aliquot containing 5 µg of protein was incubated with 1 µg of poly-dIdC in incubation buffer; then, 1 µg of 32P-labeled-DNA probe (gcg-G/A-G-T/A-A/T-ATTTcgc) was added in a total volume of 50 µl, and the incubation was allowed to continue for another 30 min at 25°C. Bound and free probes were separated by 4% polyacrylamide gel electrophoresis in buffer (pH 8.5) containing 50 mM Tris, 0.38 M glycine and 2 mM EDTA at a constant voltage of 11 V/cm for 1.5 h at 4°C. Dried gels were analyzed by autoradiography.

Supporting Information

Figure S1.

Direct sequencing data after SELEX assay.

https://doi.org/10.1371/journal.pone.0008004.s001

(3.58 MB TIF)

Table S1.

Hit gene promoters of each candidate gene.

https://doi.org/10.1371/journal.pone.0008004.s002

(0.32 MB PDF)

Author Contributions

Conceived and designed the experiments: TM TK MT. Performed the experiments: TM. Analyzed the data: TM. Contributed reagents/materials/analysis tools: TM. Wrote the paper: TM.

References

  1. 1. Reeves R, Beckerbauer L (2001) HMGI/Y proteins: flexible regulators of transcription and chromatin structure. Biochim Biophs Acta 1519: 13–29.
  2. 2. Reeves R (2001) Molecular biology of HMGA proteins: hubs of nuclear function. Gene 277: 63–81.
  3. 3. Choi1 D, Appukuttan B, Binek SJ, Planck SR, Stout JT, et al. (2008) Prediction of cis-Regulatory elements controlling genes differentially expressed by retinal and choroidal vascular endothelial cells. J Ocul Biol Dis Infor 1: 37–45.
  4. 4. Treff NR, Dement GA, Adair JE, Britt RL, Nie R, et al. (2004) Human KIT ligand promoter is positively regulated by HMGA1 in breast and ovarian cancer cells. Oncogene 23: 8557–8562.
  5. 5. Adair E, Maloney SC, Dement GA, Wertzler KJ, Smerdon MJ, et al. (2007) High-mobility group A1 proteins inhibit expression of nucleotide excision repair factor xeroderma pigmentosum group A. Cancer Res 67: 6044–6052.
  6. 6. Tesfaye A, Di Cello F, Hillion J, Ronnett BM, Elbahloul O, et al. (2007) The high-mobility group A1 gene up-regulates cyclooxygenase 2 expression in uterine tumorigenesis. Cancer Res 67: 3998–4004.
  7. 7. Ji YS, Xu Q, Schmedtje JF Jr (1998) Hypoxia induces high-mobility-group protein I(Y) and transcription of the cyclooxygenase-2 gene in human vascular endothelium. Circ Res 83: 295–304.
  8. 8. Bonnefoy E, Bandu MT, Doly J (1999) Specific binding of high-mobility-group I (HMGI) protein and histone H1 to the upstream AT-rich region of the murine beta interferon promoter: HMGI protein acts as a potential antirepressor of the promoter. Mol Cell Biol 19: 2803–2816.
  9. 9. Lin SC (2006) Identification of an NF-Y/HMG-I(Y)-binding site in the human IL-10 promoter. Mol Immunol 43: 1325–1331.
  10. 10. Chuvpilo S, Schomberg C, Gerwig R, Heinfling A, Reeves R, et al. (1993) Multiple closely-linked NFAT/octamer and HMG I(Y) binding sites are part of the interleukin4 promoter. Nucleic Acids Res 21: 5694–5704.
  11. 11. Perrella MA, Pellacani A, Wiesel P, Chin MT, Foster LC, et al. (1999) High mobility group-I(Y) protein facilitates nuclear factor-kappaB binding and transactivation of the inducible nitric-oxide synthase promoter/enhancer. J Biol Chem 274: 9045–9052.
  12. 12. Chin MT, Pellacani A, Wang H, Lin SSJ, Jain MK, et al. (1998) Enhancement of serum-response factor-dependent transcription and DNA binding by the architectural transcription factor HMG-I(Y). J Biol Chem 273: 9755–9760.
  13. 13. Treff NR, Pouchnik D, Dement GA, Britt RL, Reeves R (2004) High-mobility group A1a protein regulates Ras/ERK signaling in MCF-7 human breast cancer cells. Oncogene 23: 777–785.
  14. 14. Takamiya R, Baron RM, Yet SF, Layne MD, Perrella MA (2008) High mobility group A1 protein mediates human nitric oxide synthase 2 gene expression. FEBS Lett 582: 810–814.
  15. 15. Okuda H, Manabe T, Yanagita T, Matsuzaki S, Bando Y, et al. (2006) Novel interaction between HMGA1a and StIP1 in murine terminally differentiated retina. Mol Cell Neurosci 33: 81–87.
  16. 16. Manabe T, Katayama T, Sato N, Gomi F, Hitomi J, et al. (2003) Induced HMGA1a expression causes aberrant splicing of Presenilin-2 pre-mRNA in sporadic Alzheimer's disease. Cell Death Differ 10: 698–708.
  17. 17. Yanagita T, Manabe T, Okuda H, Matsuzaki S, Bando Y, et al. (2005) Possible involvement of the expression and phosphorylation of N-Myc in the induction of HMGA1a by hypoxia in the human neuroblastoma cell line. Neurosci Lett 374: 47–52.
  18. 18. Chau KY, Keane-Myers AM, Fedele M, Ikeda Y, Creusot RJ, et al. (2005) IFN-gamma gene expression is controlled by the architectural transcription factor HMGA1. Int Immunol 17: 297–306.
  19. 19. Fedele M, Fidanza V, Battista S, Pentimalli F, Klein-Szanto AJ, et al. (2006) Haploinsufficiency of the Hmga1 gene causes cardiac hypertrophy and myelo-lymphoproliferative disorders in mice. Cancer Res 66: 2536–2543.
  20. 20. Himes SR, Coles LS, Reeves R, Shannon MF (1996) High mobility group protein I(Y) is required for function and for c-Rel binding to CD28 response elements within the GM-CSF and IL-2 promoters. Immunity 5: 479–489.
  21. 21. Chau KY, Munshi N, Keane-Myers A, Cheung-Chau KW, Tai AK, et al. (2000) The architectural transcription factor high mobility group I(Y) participates in photoreceptor-specific gene expression. J Neurosci 20: 7317–7324.
  22. 22. Foster LC, Wiesel P, Huggins GS, Pañares R, Chin MT, et al. (2000) Role of activating protein-1 and high mobility group-I(Y) protein in the induction of CD44 gene expression by interleukin-1beta in vascular smooth muscle cells. FASEB J 14: 368–378.
  23. 23. De Martino I, Visone R, Wierinckx A, Palmieri D, Ferraro A, et al. (2009) HMGA proteins up-regulate CCNB2 gene in mouse and human pituitary adenomas. Re Cancer s 69: 1844–1850.
  24. 24. Duncan B, Zhao K (2007) HMGA1 mediates the activation of the CRYAB promoter by BRG1. DNA Cell Biol 26: 745–752.
  25. 25. Chiappetta G, Botti G, Monaco M, Pasquinelli R, Pentimalli F, et al. (2004) HMGA1 protein overexpression in human breast carcinomas: correlation with ErbB2 expression. Clin Cancer Res 10: 7637–7644.
  26. 26. Chase MB, Haga SB, Hankins WD, Williams DM, Bi Z, et al. (1999) Binding of HMG-I(Y) elicits structural changes in a silencer of the human beta-globin gene. Am J Hematol 60: 27–35.
  27. 27. Abdulkadir SA, Krishna S, Thanos D, Maniatis T, Strominger JL, et al. (1995) Functional roles of the transcription factor Oct-2A and the high mobility group protein I/Y in HLA-DRA gene expression. J Exp Med 182: 487–500.
  28. 28. Liau SS, Rocha F, Matros E, Redston M, Whang E (2008) High mobility group AT-hook 1 (HMGA1) is an independent prognostic factor and novel therapeutic target in pancreatic adenocarcinoma. Cancer 113: 302–314.
  29. 29. Manabe T, Ohe K, Katayama T, Matsuzaki S, Yanagita T, et al. (2007) HMGA1A: Sequence-specific RNA-binding factor causing sporadic Alzheimer's disease-linked exon skipping of presenilin-2 pre-mRNA. Genes Cells 12: 1179–1191.
  30. 30. Miyake K, Yoshizumi T, Imura S, Sugimoto K, Batmunkh E, et al. (2008) Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas 36: e1–9.
  31. 31. Sawai H, Yamamoto M, Okada Y, Sato M, Akamo Y, et al. (2001) Alteration of integrins by interleukin-1alpha in human pancreatic cancer cells. Pancreas 23: 399–405.
  32. 32. Baran B, Bechyne I, Siedlar M, Szpak K, Mytar B, et al. (2009) Blood monocytes stimulate migration of human pancreatic carcinoma cells in vitro: The role of tumour necrosis factor - alpha. Eur J Cell Biol. In press. DOI:https://doi.org/10.1016/j.ejcb.2009.08.002.
  33. 33. Heaney AP, Horwitz GA, Wang Z, Singson R, Melmed S (1999) Early involvement of estrogen-induced pituitary tumor transforming gene and fibroblast growth factor expression in prolactinoma pathogenesis. Nat Med 5: 1317–1321.
  34. 34. Schmutzler C, Mentrup B, Schomburg L, Hoang-Vu C, Herzog V, et al. (2007) Selenoproteins of the thyroid gland: expression, localization and possible function of glutathione peroxidase 3. Biol Chem 388: 1053–1059.
  35. 35. Wascher RA, Bostick PJ, Huynh KT, Turner R, Qi K, et al. (2001) Detection of MAGE-A3 in breast cancer patients' sentinel lymph nodes. Br J Cancer 85: 1340–1346.
  36. 36. Kondo T, Zhu X, Asa SL, Ezzat S (2007) The cancer/testis antigen melanoma-associated antigen-A3/A6 is a novel target of fibroblast growth factor receptor 2-IIIb through histone H3 modifications in thyroid cancer. Clin Cancer Res 13: 4713–4720.
  37. 37. Oh KB, Stanton MJ, West WW, Todd GL, Wagner KU (2007) Tsg101 is upregulated in a subset of invasive human breast cancers and its targeted overexpression in transgenic mice reveals weak oncogenic properties for mammary cancer initiation. Oncogene 26: 5950–5959.
  38. 38. Bai Z, Gust R (2009) Breast cancer, estrogen receptor and ligands. Arch Pharm (Weinheim) 342: 133–149.
  39. 39. Mayer IA (2009) Treatment of HER2-positive metastatic breast cancer following initial progression. Clin Breast Cancer Suppl 2: S50–7.
  40. 40. Götte M, Yip GW (2006) Heparanase, hyaluronan, and CD44 in cancers: a breast carcinoma perspective. Cancer Res 66: 10233–10237.
  41. 41. Najy AJ, Day KC, Day ML (2008) The ectodomain shedding of E-cadherin by ADAM15 supports ErbB receptor activation. J Biol Chem 28: 18393–18401.
  42. 42. Sansone P, Storci G, Tavolari S, Guarnieri T, Giovannini C, et al. (2007) IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest 117: 3988–4002.
  43. 43. Fay J, Kelehan P, Lambkin H, Schwartz S (2009) Increased expression of cellular RNA-binding proteins in HPV-induced neoplasia and cervical cancer. J Med Virol 81: 897–907.
  44. 44. Sasaki M, Nakahira K, Kawano Y, Katakura H, Yoshimine T, et al. (2001) MAGE-E1, a new member of the melanoma-associated antigen gene family and its expression in human glioma. Cancer Res 61: 4809–4814.
  45. 45. Kim ES, Khuri FR, Herbst RS (2001) Epidermal growth factor receptor biology (IMC-C225). Curr Opin Oncol 13: 506–513.
  46. 46. Onguru O, Scheithauer BW, Kovacs K, Vidal S, Jin L, et al. (2004) Analysis of epidermal growth factor receptor and activated epidermal growth factor receptor expression in pituitary adenomas and carcinomas. Mod Pathol 17: 772–780.
  47. 47. Long JL, Strocker AM, Wang MB, Blackwell KE (2009) EGFR expression in primary squamous cell carcinoma of the thyroid. Laryngoscope 119: 89–90.
  48. 48. Ebert MP, Hernberg S, Fei G, Sokolowski A, Schulz HU, et al. (2001) Induction and expression of cyclin D3 in human pancreatic cancer. J Cancer Res Clin Oncol 127: 449–454.
  49. 49. Saeger W, Schreiber S, Lüdecke DK (2001) Cyclins D1 and D3 and topoisomerase II alpha in inactive pituitary adenomas. Endocr Pathol 12: 39–47.
  50. 50. Russell A, Thompson MA, Hendley J, Trute L, Armes J, et al. (1999) Cyclin D1 and D3 associate with the SCF complex and are coordinately elevated in breast cancer. Oncogene, 18: 1983–1991.
  51. 51. Baldassarre G, Belletti B, Bruni P, Boccia A, Trapasso F, et al. (1999) Overexpressed cyclin D3 contributes to retaining the growth inhibitor p27 in the cytoplasm of thyroid tumor cells. J Clin Invest 104: 865–874.
  52. 52. Zhou R, Shanas R, Nelson MA, Bhattacharyya A, Shi J (2009) Increased expression of the heterogeneous nuclear ribonucleoprotein K in pancreatic cancer and its association with the mutant p53. Int J Cancer. In press. DOI:https://doi.org/10.1002/ijc.24744.
  53. 53. Kappeler L, Gourdji D, Zizzari P, Bluet-Pajot MT, Epelbaum J (2003) Age-associated changes in hypothalamic and pituitary neuroendocrine gene expression in the rat. J Neuroendocrinol 15: 592–601.
  54. 54. Mandal M, Vadlamudi R, Nguyen D, Wang RA, Costa L, et al. (2001) Growth factors regulate heterogeneous nuclear ribonucleoprotein K expression and function. J Biol Chem 276: 9699–9704.
  55. 55. Gorla L, Cantù M, Miccichè F, Patelli C, Mondellini P, et al. (2006) RET oncoproteins induce tyrosine phosphorylation changes of proteins involved in RNA metabolism. Cell Signal 18: 2272–2282.