GET THE APP

UHRF1: A Diagnostic and Prognostic Marker of Cancer
..

Molecular Biology: Open Access

ISSN: 2168-9547

Open Access

Research Article - (2023) Volume 12, Issue 3

UHRF1: A Diagnostic and Prognostic Marker of Cancer

Qurat-ul-Ain1*, Mehak Nimra2, Areej Ghouri3, Shafaq Zahoor4 and Asma Sarfraz5
*Correspondence: Qurat-ul-Ain, Hamdard University, Islamabad, Pakistan, Email:
1Hamdard University, Islamabad, Pakistan
2National Institute of Health, Islamabad, Pakistan
3Quaid-e-Azam University, Islamabad, Pakistan
4Riphah International University, Islamabad, Pakistan
5Shifa Tameer e Millat University, Islamabad, Pakistan

Received: 29-May-2023, Manuscript No. MBL-23-100498; Editor assigned: 30-May-2023, Pre QC No. P-100498; Reviewed: 12-Jun-2023, QC No. Q-100498; Revised: 17-Jun-2023, Manuscript No. R-100498; Published: 26-Jun-2023 , DOI: 10.37421/2168-9547.2023.12.376
Citation: Ain, Qurat-Ul, Mehak Nimra, Areej Ghouri and Shafaq Zahoor, et al. “UHRF1: A Diagnostic and Prognostic Marker of Cancer.” Mol Bio 12 (2023): 376.
Copyright: © 2022 Ain QU, et al. This is an open-access article distributed under the terms of the creative commons attribution license which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

Introduction

There are many members of the UHRF family including UHRF1, UHRF2, UHRF3 and UHRF4 having different functions. In this review, we mainly study the activity of UHRF1. It is a multi-domain protein associated with epigenetic mechanisms of cell regulation and proliferation. UHRF1 stands for Ubiquitinlike PHD Ring finger 1 having a location in the chromosomal region 19p13.3 [1]. It plays a key role in transferring methylation from mother to daughter DNA strands [2-5]. mUHRF1 was discovered against murine thymic lymphoma by engineering antibodies. hUHRF1 has activity of E3 ligases for histones H3. Expression of UHRF1 increases in breast cancer, cervical lesions, rhabdomyosarcoma, pancreatic adenocarcinoma, prostate cancer and lung cancer [6]. Increased expression of UHRF1 in human pulmonary fibroblasts causes increased topoisomerase IIα expression and hence increased cell proliferation. Similarly, depletion of UHRF1 causes DNA damage response, G2/M phase arrest and apoptosis formation [7-9]. UHRF1 by interacting with DNMT1 and HDAC1 induces heterochromatin structure.

Discussion

Structure of UHRF1

It possesses a UBL (Ubiquitin-like domain), TTD (Cryptic Tandem Tudor Domain), PHD (Plant Homeodomain), SRA (Set and Ring Associated) and RING (Really Interesting New Gene) domain [10] as shown in (Figure 1).

molecular-biology-proteins

Figure 1.Construction of UHRF1: UBL (Ubiquitin-Like Domain), TTD (Cryptic Tandem Tudor Domain), PHD (Plant Homeodomain), SRA (Set and Ring Associated) and RING (Really Interesting New Gene) domains interact with DNMT1, HDAC1, H3K9 and Histone H3 proteins that lead to epigenetics code of inheritance.

UBL domain contains α/β ubiquitin folds with surface Lysine residues i.e. 35% similar to ubiquitin, consisting of 76 amino acids and has an important role in cell cycle progression, protein degradation and gene transcription [11]. PH Domain recognizes di and tri-methylation of histone h3 lysine 9 (H3K9), associated with heterochromatin formation, transcriptional processes and also with downregulation of UHRF1 both in human and mouse causes disrupt H3K9 distribution. PHD promotes gene activation and inactivation by interacting with tri-methylated H3K4. Various studies have shown that PHD has the ability to read the histone code as well [12]. The SRA domain contains 150-170 amino acids directly involved in DNA methylation to the target sequences by recognizing hemimethylated Cytosine of new daughter DNA strand [13] binds with DNMT1 and involved in heterochromatic formation along with PH domain. It sets a bridge between DNA methylation and histone code by allowing UHRF1 to bind with HDAC1, methylated DNA and DNMT1. RING is 76 amino acid long polypeptide domain, attached with Lysine on cellular protein by its C-terminal. Ubiquitination is mediated by E1/E2/E3 enzymes; especially E3 catalyzes the binding of the C-terminal with Lysine on targeted protein. There are two main classes of E3 ligases, HECT and Ring class having Ring finger domain [14]. UHRF family has auto-ubiquitinating activity [8] like many ligases which contain Ring finger domain. UHRF1 ubiquitinates histone H3 and these are substrates for the UHRF family.

Mechanism of UHRF1 in heterochromatin formation

• First UHRF1 binds to PCNA and recognizes hemimethylated DNA through its SRA domain DNA replication occurs and histones are reassembled immediately on the DNA strand shown in step 1 Figure 2 [1,4,5].

• Secondly UHRF1 recruits the G9a which methylates the h3k9 (Histone h3 Lysine 9) and methylated h3k9 binds with the PHD domain of the UHRF1 Step 2 in figure 7 then it recruits the DNMT1which methylates both DNA strands and transfers the methylation status from mother to daughter DNA strand.

• Finally UHRF1 recruits the histone deacetylase 1 (HDAC1) which deacetylates the histone proteins and helps in heterochromatin formation and transcriptional suppression [15] Step 3 in (Figure 2).

molecular-biology-methylates

Figure 2. Mechanism of UHRF1 in heterochromatin formation. UHRF1 binds to PCNA and recognizes hemimethylated DNA through its SRA domain. UHRF1 recruits the G9a, DNMT1 and HDAC1 which methylates the h3k9 and binds with PHD domain, both DNA strands and transfer the methylation status from mother to daughter DNA strand and deacetylates the histone proteins respectively which leads to heterochromatin formation.

UHRF1 and epigenetic codes

UHRF members directly influence the histone code by their enzymatic activity (E3 Ligase) via RING, maintaining the epigenetic code (DNA Methylation and Histone Code) and genomic integrity. It might be a tumor suppressor gene [16].

UHRF1 interacts with the methyl-CpG region in DNA strand, methylated H3K9, DNMT1, HDAC1, PCNA and G9a, links with DNA methylation and histone methylation, deacetylation and ubiquitination with heterochromatin formation as shown in Figure 3 [2,4,5,7,10,15,17]. DNA methylation and histone modification both act together, changing the gene expression and heterochromatin structure [4,18,19] (Figure 3).

molecular-biology-methylation

Figure 3. UHRF1 links with DNA methylation, histone methylation, deacetylation and ubiquitination, changes the gene expression and heterochromatin formation.

Up regulation of UHRF1

UHRF1 is significantly over-expressed in various cancers and tumor cells [8,20]. Upregulation of UHRF1 is associated with high levels of p73, SIRT1, Caspase 3, DNMT1 and HDAC1 level and ultimately leads to cancer as shown in Figure 4. UHRF1 when associated with DNMT1 maintains the methylation status of daughter DNA and maintains the epigenetic inheritance [2,21]. UHRF1 interacts with HDAC1 and transfers them to the methylated tumor suppressor genes [20]. It also maintains histone deacetylation and histone methylation [22] and plays many roles in cancer proving it beneficial for therapeutic targeting [1,23] (Figure 4).

molecular-biology-regulation

Figure 4. Upstream regulation of UHRF1: Upregulation of UHRF1 is associated with high levels of p73, SIRT1, Caspase 3, DNMT1 and HDAC1 level and ultimately leads to cancer.

Downregulation of UHRF1

Down regulation of UHRF1 causes DNA damage by breaking DNA strands [24,25], cell cycle arrest in the G2/M stage by inhibiting Cyclin-Dependent Kinases 1 (cdk-1), a regulator of cell cycle progression in G2 phase during mitosis and apoptosis by activating Caspase 8 [9] as shown in Figure 5. chk 1 and chk 2 are activated by phosphorylation in DNA damage response. Loss of chk 2 occurs in UHRF1-depleted cells and inhibits the phosphorylation of chk 1, it leads to cell death by cell cycle arrest. Apoptosis results from depletion of UHRF1 are p53 independent and this pathway regulates Caspase 8 functions which causes activation of Caspase 3 and ultimately apoptosis [26]. Various studies have shown that depletion of UHRF1 prevents cell cycle progression so it is useful to prevent the growth of tumor cells in cancer treatment [27] and UHRF1 depleted cells become more sensitive to DNA damaging agents [16,28] (Figure 5).

molecular-biology-depletion

Figure 5. Depletion of UHRF1: Down-regulation of UHRF1 causes DNA damage by breaking DNA strands, cell cycle arrest in the G2/M stage by inhibiting CDK-1 and apoptosis by activating Caspase 8.

Role of UHRF1 in cancers

UHRF1 is down regulated by p53 via the up-regulation of p21 and deactivation of E2F1, an up-regulator of UHRF1 [7,16,20]. As p53 is deficient in 50% of cancers [29], So UHRF1 is up regulated in many cancers by the following mechanism. In p53 deficient cancers, the cyclin D/cdks (Cyclin- Dependent Kinases) complex become activated, causing phosphorylation of PRB and phosphorylated PRB activates the E2F1 which binds with UHRF1 and up regulates its level which leads to cancer as shown in Figure 6. UHRF1 is up regulated by rapid cell cycle progression as well [20,7,8,30,31]. In the rapid cell cycle, UHRF1 binds with newly synthesized DNA with PCNA, DNMT1 and HDAC1 and hastily transfers the CH3 group from mother to daughter progeny. It activates the G1/S phase and the cyclin B/cdk1 complex becomes activated8 in the G2/M phase and p21 inactivates this cdk1. [32] (Figure 6).

molecular-biology-cancer

Figure 6. Role of UHRF1 in cancer.

UHRF1 as a diagnostic and prognostic marker

Up-regulation of UHRF1 has been associated with various cancers e.g, including breast cancer, lungs cancer, bladder, prostate, cervical and pancreatic cancers [8,20,30,31,33,34]. Up-regulation of UHRF1 is associated with the downregulation of p53 as it has already been discussed in the previous paragraph and it is also essential for cell cycle progression so UHRF1 down-regulation is associated with cell growth suppression. It has been used as a diagnostic or prognostic marker by quantitative analysis of UHRF1 in urine and tissue samples of different cancer patients. UHRF1 and other proteins’ expression can be detected by immune histochemistry [7].

UHRF1 as a potential therapeutic target

All UHRF1 members have ubiquitin ligase E3 activity, targeting E3 ligases proved beneficial as it is an ideal drug target in anticancer therapy. Expression of E3 ligases along with UHRF1 increases in cancer cells so when they are inhibited, growth arrest and apoptosis occur [35,36]. So, targeting UHRF1 is selective anticancer therapy.

DNA methylation pattern in UHRF1

The most important domain involved in DNA methylation is the UHRF1-SRA domain. UHRF1 binds with hemimethylated DNA with the SRA domain which is involved in the proper setting of DNMT1 on the DNA strand [4,6]. DNMT1 and UHRF1 are two proteins that have affinity and selectivity for hemimethylated DNA on their own and both are essential for performing their functions [37]. It is supposed that UHRF1 moves along DNA strands, recognizes hemimethylated CpG region [3-5] and it dictates DNMT1 through an unknown mechanism to catalyze methylation at the target site. Another model states that when DNMT1 starts acting, UHRF 1 gets separated from the CpG region, allowing DNMT1 to work properly. It is widely researched how DNA hypermethylation makes the gene transcriptionally silent. DNA methylation behaves as a signal for the recruitment of CpG Methyl Binding Domain (MBD) [38] which further recruits the Histone Deacetylase (HDAC) and form many genes silencing complexes as shown in (Figure 7).

molecular-biology-target

Figure 7. DNA methylation pattern in UHRF1: UHRF1 moves along DNA strands, recognizes hemimethylated CpG region and dictates DNMT1 to catalyze methylation at the target site. DNA methylation behaves as a signal for the recruitment of CpG region MBD which further recruits HDAC1 and form many genes silencing complexes.

Factor influencing DNA methylation

Many factors have a direct impact on the extent of DNA methylation pattern.

Aging: As the tissue becomes aged, there is more possibility that the genome will become hypomethylated and certain CpG islands become hypermethylated. But it is not known whether this change makes the person more susceptible to cancer or not [39].

Diet: DNA methylation requires methyl group which comes from folate and methionine components of the diet. As mammals lack the ability to produce folate and methionine themselves, they are totally dependent on diet for carrying out DNA methylation. A diet containing a low amount of these substances causes decrease DNA methylation and increases the tendency of cancer [40-43].

Environment: DNA methylation is also affected by agents such as Arsenic and Cadmium. Ras gene hypomethylation is caused by Arsenic and total DNA hypomethylation is caused by Cadmium which does so by inactivating DNMT1 [44-49].

UHRF1 is a drug able to target cancer therapy

There are 3 ways by which UHRF1 acts as drug able target in cancer therapy;

• Inhibition of UHRF1 expression inhibits the cells from entering into the S phase and thus it causes growth arrest. Over-expression of UHRF1 overcomes cell contact inhibition in human lung fibroblasts. Down regulation of UHRF1 activates the DNA damage response and causes cell arrest and apoptosis [50-54].

• UHRF1 members are ubiquitin E3 ligases and inhibiting the proteosomes pathway is one of the strategies in anticancer drug development.

• E3 ligases are over-expressed in many cancers and inhibition of E3 ligases causes the growth inhibition of cells and apoptosis in cancer cells. So, E3 ligases can also be targeted to avoid unwanted effects [55-62].

It would be interesting to find a direct inhibitor of UHRF1, it would be helpful because it directly inhibits the cancer cells’ growth and causes apoptosis. Therefore, research on the direct inhibition of SRA and RING domain with its E3 ligase activity is under consideration [63-68].

Conclusion

UHRF1 is an attractive potential therapeutic target

It has been shown that with chemical stimulation of double-stranded breaks in DNA by an anticancer agent (Adriamycin) in HCT116 cells (colon cancer) there is a decrease in UHRF1 expression at the transcriptional level and protein. This fall, required for the cells moving towards apoptosis, is controlled by the way p53/p21WAF1/CIP1. By cons, when p53 is defective, there is no decline in the expression of UHRF1. Consequently, one might consider that the lack of decrease in the expression and/or over-expression of UHRF1 in cancer cells results from an alteration of the p53 tumor suppressor gene.

When interferes with the ubiquitin ligase activity of UHRF1 (by overexpression of RING domain mutant sound) increases the sensitivity of cancer cells to chemotherapeutic agents. In addition, it was reported that inhibition of the expression of UHRF1 causes a reduction of Ribonucleotide Reductase, an enzyme essential for the Synthesis of Deoxy Nucleotides. The concomitant decrease in the expression of both proteins leads to increased cell sensitivity to hydroxyurea (HydréaTM), which can be particularly effective in the treatment of leukemia. It was suggested that the anti-transcriptional targeting of UHRF1 might be interesting in the case of cancers resistant to hydroxyurea without resorting to the increase in therapeutic doses.

All these studies seem to converge on the fact that UHRF1 is an attractive target to develop new anti-cancer molecules.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Unoki, Motoko. "Current and potential anticancer drugs targeting members of the UHRF1 complex including epigenetic modifiers." Recent Pat Anticancer Drug Discov 6 (2011): 116-130.
  2. Google Scholar, Crossref, Indexed at

  3. Sharif, Jafar, Masahiro Muto, Shin-ichiro Takebayashi and Isao Suetake, et al. "The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA."Nature450 (2007): 908-912.
  4. Google Scholar, Crossref, Indexed at

  5. Arita, Kyohei, Mariko Ariyoshi, Hidehito Tochio and Yusuke Nakamura, et al. "Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism."Nature455 (2008): 818-821.
  6. Google Scholar, Crossref, Indexed at

  7. Hashimoto, Hideharu, John R. Horton, Xing Zhang and Magnolia Bostick, et al. "The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix."Nature455 (2008): 826-829.
  8. Google Scholar, Crossref, Indexed at

  9. Avvakumov, George V., John R. Walker, Sheng Xue and Yanjun Li, et al. "Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1."Nature455 (2008): 822-825.
  10. Google Scholar, Crossref, Indexed at

  11. Bronner, Christian, Mayada Achour, Yoshimi Arima and Thierry Chataigneau, et al. "The UHRF family: oncogenes that are drugable targets for cancer therapy in the near future?." Pharmacol Ther 115 (2007): 419-434.
  12. Google Scholar, Crossref, Indexed at

  13. Unoki, Motoko, Toshihiko Nishidate and Yusuke Nakamura. "ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain."Oncogene23 (2004): 7601-7610.
  14. Google Scholar, Indexed at

  15. Jenkins, Yonchu, Vadim Markovtsov, Wayne Lang and Poonam Sharma, et al. "Critical role of the ubiquitin ligase activity of UHRF1, a nuclear RING finger protein, in tumor cell growth."Mol Biol Cell 16 (2005): 5621-5629.
  16. Google Scholar, Crossref, Indexed at

  17. Tien, Amy L., Sucharita Senbanerjee, Atul Kulkarni and Raksha Mudbhary, et al. "UHRF1 depletion causes a G2/M arrest, activation of DNA damage response and apoptosis."Biochem435 (2011): 175-185.
  18. Google Scholar, Crossref, Indexed at

  19. Achour, M., X. Jacq, P. Ronde and M. Alhosin, et al. "The interaction of the SRA domain of ICBP90 with a novel domain of DNMT1 is involved in the regulation of VEGF gene expression."Oncogene27 (2008): 2187-2197.
  20. Google Scholar, Crossref, Indexed at

  21. Sun, Yi. "Targeting E3 ubiquitin ligases for cancer therapy."Cancer Biol Med 2 (2003): 623-629.
  22. Indexed at

  23. Shi, Yigong. "Mechanisms of caspase activation and inhibition during apoptosis."Mol cell9 (2002): 459-470.
  24. Google Scholar, Crossref, Indexed at

  25. Dodge, Jonathan E., Masaki Okano, Fred Dick and Naomi Tsujimoto, et al. "Inactivation of Dnmt3b in mouse embryonic fibroblasts results in DNA hypomethylation, chromosomal instability, and spontaneous immortalization."J Biol Chem 280 (2005): 17986-17991.
  26. Google Scholar, Crossref, Indexed at

  27. Chen, Ceshi, Arun K. Seth and Andrew E. Aplin. "Genetic and expression aberrations of E3 ubiquitin ligases in human breast cancer."Mol Cancer 4 (2006): 695-707.
  28. Google Scholar, Crossref, Indexed at

  29. Kim, Jong Kyong, Pierre-Olivier Esteve, Steven E. Jacobsen and Sriharsa Pradhan. "UHRF1 binds G9a and participates in p21 transcriptional regulation in mammalian cells."Nucleic Acids Res 37 (2009): 493-505.
  30. Google Scholar, Crossref

  31. Muto, Masahiro, Yasuyoshi Kanari, Eiko Kubo and Tamami Takabe, et al. "Targeted disruption of Np95 gene renders murine embryonic stem cells hypersensitive to DNA damaging agents and DNA replication blocks."J Biol Chem 277 (2002): 34549-34555.
  32. Google Scholar, Crossref, Indexed at

  33. Karagianni, Panagiota, Larbi Amazit, Jun Qin and Jiemin Wong. "ICBP90, a novel methyl K9 H3 binding protein linking protein ubiquitination with heterochromatin formation."Mol Cell Biol 28 (2008): 705-717.
  34. Google Scholar, Crossref, Indexed at

  35. Delaval, Katia, Jérôme Govin, Frédérique Cerqueira and Sophie Rousseaux, et al. "Differential histone modifications mark mouse imprinting control regions during spermatogenesis." EMBO J26 (2007): 720-729.
  36. Google Scholar, Crossref, Indexed at

  37. Vaissière, Thomas, Carla Sawan and Zdenko Herceg. "Epigenetic interplay between histone modifications and DNA methylation in gene silencing ."Mutat Res Rev Mutat Res 659 (2008): 40-48.
  38. Google Scholar, Crossref, Indexed at

  39. Mousli, M., R. Hopfner, A. Q. Abbady, D. Monte and M. Jeanblanc, et al. "ICBP90 belongs to a new family of proteins with an expression that is deregulated in cancer cells."Br J Cancer89 (2003): 120-127.
  40. Google Scholar, Crossref, Indexed at

  41. Bostick, Magnolia, Jong Kyong Kim, Pierre-Olivier Estève and Amander Clark, et al. "UHRF1 plays a role in maintaining DNA methylation in mammalian cells."sci317 (2007): 1760-1764.
  42. Google Scholar, Crossref, Indexed at

  43. Rottach, Andrea, Carina Frauer, Garwin Pichler and Ian Marc Bonapace, et al. "The multi-domain protein Np95 connects DNA methylation and histone modification."Nucleic Acids Res Spec Publ 38 (2010): 1796-1804.
  44. Google Scholar, Crossref, Indexed at

  45. Xu, F., C. Mao, Y. Ding and C. Rui, et al. "Molecular and enzymatic profiles of mammalian DNA methyltransferases: structures and targets for drugs."Curr Med Chem 17 (2010): 4052-4071.
  46. Google Scholar, Crossref, Indexed at

  47. Mistry, Helena, Lianne Gibson, Ji Weon Yun and Haya Sarras, et al. "Interplay between Np95 and Eme1 in the DNA damage response."Biochem Biophys Res Commun375 (2008): 321-325.
  48. Google Scholar, Crossref, Indexed at

  49. Mistry, Helena, Laura Tamblyn, Hussein Butt and Daniel Sisgoreo, et al. "UHRF1 is a genome caretaker that facilitates the DNA damage response to γ-irradiation."Genome Integr 1 (2010): 1-12.
  50. Google Scholar, Crossref, Indexed at

  51. Afshar, Golnar, Nannette Jelluma, Xiaodong Yang and Daniel Basila, et al. "Radiation-induced caspase-8 mediates p53-independent apoptosis in glioma cells." Cancer Res 66 (2006): 4223-4232.
  52. Google Scholar, Crossref, Indexed at

  53. Morgan, David O. "Cyclin-dependent kinases: engines, clocks and microprocessors."Annu Rev Cell Dev Biol13 (1997): 261-291.
  54. Google Scholar, Crossref, Indexed at

  55. Arima, Yoshimi, Toru Hirota, Christian Bronner and Marc Mousli, et al. "Down‐regulation of nuclear protein ICBP90 by p53/p21Cip1/WAF1‐dependent DNA‐damage checkpoint signals contributes to cell cycle arrest at G1/S transition."Genes Cells 9 (2004): 131-142.
  56. Google Scholar, Crossref, Indexed at

  57. Hussain, S. Perwez and Curtis C. Harris. "Molecular epidemiology and carcinogenesis: endogenous and exogenous carcinogens."Mutat Res 462 (2000): 311-322.
  58. Google Scholar, Crossref, Indexed at

  59. Oba-Shinjo, Sueli M., Mario H. Bengtson, Sheila MB Winnischofer and Christian Colin, et al. "Identification of novel differentially expressed genes in human astrocytomas by cDNA representational difference analysis."Mol Brain 140 (2005): 25-33.
  60. Google Scholar, Crossref, Indexed at

  61. Crnogorac–Jurcevic, Tatjana, Rathi Gangeswaran, Vipul Bhakta and Gabriele Capurso, et al. "Proteomic analysis of chronic pancreatitis and pancreatic adenocarcinoma."Gastroenterology129 (2005): 1454-1463.
  62. Google Scholar, Crossref, Indexed at

  63. Brunet, Julie, Alexander W. Pfaff, Ahmed Abidi and Motoko Unoki, et al. "Toxoplasma gondii exploits UHRF1 and induces host cell cycle arrest at G2 to enable its proliferation."Cell Microbiol 10 (2008): 908-920.
  64. Google Scholar, Crossref, Indexed at

  65. Lorenzato, Marianne, Stephanie Caudroy, Christian Bronner and Ghislaine Evrard, et al. "Cell cycle and/or proliferation markers: what is the best method to discriminate cervical high-grade lesions?."Hum Pathol 36 (2005): 1101-1107.
  66. Google Scholar, Crossref, Indexed at

  67. Pita, Jaime Miguel, A. Banito, Branca Maria Cavaco and Valeriano Leite. "Gene expression profiling associated with the progression to poorly differentiated thyroid carcinomas."Br J Cancer 101 (2009): 1782-1791.
  68. Google Scholar

  69. Chen, Wen Yong, David H. Wang, RayWhay Chiu Yen and Jianyuan Luo, et al. "Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses."Cell123 (2005): 437-448.
  70. Google Scholar, Crossref, Indexed at

  71. Sun, Yi. "E3 ubiquitin ligases as cancer targets and biomarkers."Neoplasia8 (2006): 645-654.
  72. Google Scholar, Crossref, Indexed at

  73. Bronner, Christian, Guy Fuhrmann, Frédéric L. Chédin and Marcella Macaluso, et al. "UHRF1 links the histone code and DNA methylation to ensure faithful epigenetic memory inheritance."Genet Epigenetics2 (2009): GEG-S3992.
  74. Google Scholar, Crossref, Indexed at

  75. Jones, Peter A. and Stephen B. Baylin. "The epigenomics of cancer."Cell128 (2007): 683-692.
  76. Google Scholar, Crossref, Indexed at

  77. Momparler, Richard L. "Cancer epigenetics."Oncogene22 (2003): 6479-6483.
  78. Google Scholar, Crossref, Indexed at

  79. Lund, Anders H. and Maarten van Lohuizen. "Epigenetics and cancer."Genes Dev 18 (2004): 2315-2335.
  80. Google Scholar, Crossref, Indexed at

  81. Bestor, Timothy H. "Activation of mammalian DNA methyltransferase by cleavage of a Zn binding regulatory domain."EMBO J 11 (1992): 2611-2617.
  82. Google Scholar, Crossref, Indexed at

  83. Bird, Adrian P. "CpG-rich islands and the function of DNA methylation."Nature321 (1986): 209-213.
  84. Google Scholar, Crossref, Indexed at

  85. Bonfils, Claire, Normand Beaulieu, Eric Chan and Judith Cotton-Montpetit, et al. "Characterization of the human DNA methyltransferase splice variant Dnmt1b."J Biol Chem 275 (2000): 10754-10760.
  86. Google Scholar, Crossref, Indexed at

  87. Buschhausen, G., B. Wittig, M. Graessmann and A. Graessmann. "Chromatin structure is required to block transcription of the methylated herpes simplex virus thymidine kinase gene."Proc Natl Acad Sci84 (1987): 1177-1181.
  88. Google Scholar, Crossref, Indexed at

  89. Wang, Chenguang, Maufu Fu, Sridhar Mani and Scott Wadler, et al. "Histone acetylation and the cell-cycle in cancer."Front Biosci 6 (2001): 610-629.
  90. Google Scholar

  91. Hans, Fabienne and Stefan Dimitrov. "Histone H3 phosphorylation and cell division."Oncogene20 (2001): 3021-3027.
  92. Google Scholar, Crossref, Indexed at

  93. Fraga, Mario F., Esteban Ballestar, Ana Villar-Garea and Manuel Boix-Chornet, et al. "Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer."Nat Genet 37 (2005): 391-400.
  94. Google Scholar, Crossref, Indexed at

  95. Glaser, Keith B., Junling Li, Michael J. Staver and Ru-Qi Wei, et al. "Role of class I and class II histone deacetylases in carcinoma cells using siRNA."Biochem Biophys Res Commun310 (2003): 529-536.
  96. Google Scholar, Crossref, Indexed at

  97. Gregoretti, IvanV, Yun-Mi Lee and Holly V. Goodson. "Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis."J Mol Biol338 (2004): 17-31.
  98. Google Scholar, Crossref, Indexed at

  99. Halkidou, Kalipso, Luke Gaughan, Susan Cook and Hing Y. Leung,et al. "Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer." Prostate59 (2004): 177-189.
  100. Google Scholar, Crossref, Indexed at

  101. Herman, James G. and Stephen B. Baylin. "Gene silencing in cancer in association with promoter hypermethylation."N Engl J Med 349 (2003): 2042-2054.
  102. Google Scholar, Crossref, Indexed at

  103. Tsai, Hsing-Chen and Stephen B. Baylin. "Cancer epigenetics: linking basic biology to clinical medicine."Cell Res21 (2011): 502-517.
  104. Google Scholar, Crossref

  105. Kim, Jin‐Hyun, Yang‐Kyu Choi, Ho‐Jeong Kwon and Han‐Kwang Yang, et al. "Downregulation of gelsolin and retinoic acid receptor β expression in gastric cancer tissues through histone deacetylase 1."Gastroenterol Hepatol19 (2004): 218-224.
  106. Google Scholar, Crossref, Indexed at

  107. Lagger, Gerda, Dónal O'Carroll, Martina Rembold and Harald Khier, et al. "Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression."EMBO J21 (2002): 2672-2681.
  108. Google Scholar, Crossref, Indexed at

  109. Launay, Sophie, Olivier Hermine, Michaela Fontenay and Guido Kroemer, et al. "Vital functions for lethal caspases."Oncogene24 (2005): 5137-5148.
  110. Google Scholar, Crossref, Indexed at

  111. Levenson, Jonathan M. and J. David Sweatt. "Epigenetic mechanisms in memory formation."Nat Rev Neurosci6 (2005): 108-118.
  112. Google Scholar, Crossref, Indexed at

  113. Lister, Ryan, Mattia Pelizzola, Robert H. Dowen and R. David Hawkins, et al. "Human DNA methylomes at base resolution show widespread epigenomic differences."nature462 (2009): 315-322.
  114. Google Scholar, Crossref, Indexed at

  115. Okano, Masaki, Daphne W. Bell, Daniel A. Haber, and En Li. "DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development."Cell99 (1999): 247-257.
  116. Google Scholar, Crossref, Indexed at

  117. Probst, Aline V., Elaine Dunleavy and Geneviève Almouzni. "Epigenetic inheritance during the cell cycle."Nat Rev Mol Cell Biol10 (2009): 192-206.
  118. Google Scholar, Crossref, Indexed at

  119. Reik, Wolf. "Stability and flexibility of epigenetic gene regulation in mammalian development."Nature447 (2007): 425-432.
  120. Google Scholar, Crossref, Indexed at

  121. Richon, Victoria M., Todd W. Sandhoff, Richard A. Rifkind and Paul A. Marks. "Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation."Proc Natl Acad Sci97 (2000): 10014-10019.
  122. Google Scholar, Crossref, Indexed at

  123. Ruthenburg, Alexander J., Haitao Li, Dinshaw J. Patel and C. David Allis. "Multivalent engagement of chromatin modifications by linked binding modules."Nat Rev Mol Cell Biol 8 (2007): 983-994.
  124. Google Scholar, Crossref, Indexed at

  125. Sambucetti, Lidia C., Denise D. Fischer, Sonya Zabludoff and Paul O. Kwon, et al. "Histone deacetylase inhibition selectively alters the activity and expression of cell cycle proteins leading to specific chromatin acetylation and antiproliferative effects."J Biol Chem274 (1999): 34940-34947.
  126. Google Scholar, Crossref, Indexed at

  127. Sasaki, Hiroyuki and Yasuhisa Matsui. "Epigenetic events in mammalian germ-cell development: reprogramming and beyond."Nat Rev Genet 9 (2008): 129-140.
  128. Google Scholar, Crossref, Indexed at

  129. Szyf, Moshe. "The DNA methylation machinery as a target for anticancer therapy."Pharmacol Ther70 (1996): 1-37.
  130. Google Scholar, Crossref, Indexed at

  131. Weake, Vikki M. and Jerry L. Workman. "Histone ubiquitination: triggering gene activity."Mol cell29 (2008): 653-663.
  132. Google Scholar, Crossref, Indexed at

  133. Weichert, Wilko, Annika Röske, Silvia Niesporek and Aurelia Noske, et al. "Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo." Clin Cancer Res 14 (2008): 1669-1677.
  134. Google Scholar, Crossref, Indexed at

  135. Zhang, Ke and Sharon YR Dent. "Histone modifying enzymes and cancer: going beyond histones."J Cell Biochem96 (2005): 1137-1148.
  136. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 607

Molecular Biology: Open Access received 607 citations as per Google Scholar report

Molecular Biology: Open Access peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward