HMGA2

Last updated
HMGA2
Identifiers
Aliases HMGA2 , BABL, HMGI-C, HMGIC, LIPO, STQTL9, high mobility group AT-hook 2, SRS5
External IDs OMIM: 600698 HomoloGene: 136767 GeneCards: HMGA2
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

RefSeq (protein)

NP_001287847
NP_001287848
NP_001317119
NP_003474
NP_003475

Contents

n/a

Location (UCSC) Chr 12: 65.82 – 65.97 Mb n/a
PubMed search [2] n/a
Wikidata
View/Edit Human

High-mobility group AT-hook 2, also known as HMGA2, is a protein that, in humans, is encoded by the HMGA2 gene. [3] [4] [5]

Function

This gene encodes a protein that belongs to the non-histone chromosomal high-mobility group (HMG) protein family. HMG proteins function as architectural factors and are essential components of the enhanceosome. This protein contains structural DNA-binding domains and may act as a transcriptional regulating factor. Identification of the deletion, amplification, and rearrangement of this gene that are associated with lipomas suggests a role in adipogenesis and mesenchymal differentiation. A gene knock-out study of the mouse counterpart demonstrated that this gene is involved in diet-induced obesity. Alternate transcriptional splice variants, encoding different isoforms, have been characterized. [5]

The expression of HMGA2 in adult tissues is commonly associated with both malignant and benign tumor formation, as well as certain characteristic cancer-promoting mutations. Homologous proteins with highly conserved sequences are found in other mammalian species, including lab mice ( Mus musculus ).

HMGA2 contains three basic DNA-binding domains (AT-hooks) that cause the protein to bind to adenine-thymine (AT)-rich regions of nuclear DNA. HMGA2 does not directly promote or inhibit the transcription of any genes, but alters the structure of DNA and promotes the assembly of protein complexes that do regulate the transcription of genes. With few exceptions, HMGA2 is expressed in humans only during early development, and is reduced to undetectable or nearly undetectable levels of transcription in adult tissues. [6] The microRNA let-7 is largely responsible for this time-dependent regulation of HMGA2. [7] The apparent function of HMGA2 in proliferation and differentiation of cells during development is supported by the observation that mice with mutant HMGA2 genes are unusually small (the pygmy or mini-mouse phenotype), [8] and genome-wide association studies linking HMGA2-associated SNPs to variation in human height. [9]

Regulation by let-7

Let-7 inhibits production of specific proteins by complementary binding to their mRNA transcripts. The HMGA2 mature mRNA transcript contains seven regions complementary or nearly complementary to let-7 in its 3' untranslated region (UTR). [10] Let-7 expression is very low during early human development, which coincides with the greatest transcription of HMGA2. The time-dependent drop in HMGA2 expression is caused by a rise in let-7 expression. [7]

Clinical significance

Relationship with cancer

Heightened expression of HMGA2 is found in a variety of human cancers, but the precise mechanism by which HMGA2 contributes to the formation of cancer is unknown. [11] [12] The same mutations that lead to pituitary adenomas in mice can be found in similar cancers in humans. [11] Its presence is associated with poor prognosis for the patient, but also with sensitization of the cancer cells to certain forms of cancer therapy. [13] To be specific, HMGA2-high cancers display an abnormally strong response to double strand breaks in DNA caused by radiation therapy and some forms of chemotherapy. Artificial addition of HMGA2 to some forms of cancer unresponsive to DNA damage cause them to respond to the treatment instead, although the mechanism by which this phenomenon occurs is also not understood. [13] However, the expression of HMGA2 is also associated with increased rates of metastasis in breast cancer, and both metastasis and recurrence of squamous cell carcinoma. These properties are responsible for patients' poor prognoses. As with HMGA2's effects on the response to radiation and chemotherapy, the mechanism by which HMGA2 exerts these effects is unknown. [13]

A very common finding in HMGA2-high cancers is the under-expression of let-7. [14] This is not unexpected, given let-7's natural role in the regulation of HMGA2. However, many cancers are found with normal levels of let-7 that are also HMGA2 high. Many of these cancers express the normal HMGA2 protein, but the mature mRNA transcript is truncated, missing a portion of the 3'UTR that contains the critical let-7 complementary regions. Without these, let-7 is unable to bind to HMGA2 mRNA, and, thus, is unable to repress it. The truncated mRNAs may arise from a chromosomal translocation that results in loss of a portion of the HMGA2 gene. [10]

ERCC1

Overexpressed HMGA2 may play a role in the frequent repression of ERCC1 in cancers. The let-7a miRNA normally represses the HMGA2 gene, and in normal adult tissues, almost no HMGA2 protein is present. [15] (See also Let-7 microRNA precursor.) Reduction or absence of let-7a miRNA allows high expression of the HMGA2 protein. As shown by Borrmann et al., [16] HMGA2 targets and modifies the chromatin architecture at the ERCC1 gene, reducing its expression. These authors noted that repression of ERCC1 (by HGMA2) can reduce DNA repair, leading to increased genome instability.

ERCC1 protein expression is reduced or absent in 84% to 100% of human colorectal cancers. [17] [18] ERCC1 protein expression was also reduced in a diet-related mouse model of colon cancer. [19] As indicated in the ERCC1 article, however, two other epigenetic mechanisms of repression of ERCC1 also may have a role in reducing expression of ERCC1 (promoter DNA methylation and microRNA repression).

Chromatin immunoprecipitation

Genome-wide analysis of HMGA2 target genes was performed by chromatin immunoprecipitation in a gastric cell line with overexpressed HMGA2, and 1,366 genes were identified as potential targets. [20] The pathways they identified as associated with malignant neoplasia progression were the adherens junction pathway, MAPK signaling pathway, Wnt signaling pathway, p53 signaling pathway, VEGF signaling pathway, Notch signaling pathway, and TGF beta signaling pathway.

Non-homologous end joining DNA repair

Overexpression of HMGA2 delayed the release of DNA-PKcs (needed for non-homologous end joining DNA repair) from double strand break sites. Overexpression of HMGA2 alone was sufficient to induce chromosomal aberrations, a hallmark of deficiency in NHEJ-mediated DNA repair. These properties implicate HMGA2 in the promotion of genome instability and tumorigenesis. [21] showed that

Base excision repair pathway

HMGA2 protein can cleave DNA containing apurinic/apyrimidinic (AP) sites (is an AP lyase). In addition, this protein also possesses the related 5’-deoxyribosyl phosphate (dRP) lyase activity. An interaction between human AP endonuclease 1 and HMGA2 in cancer cells has been demonstrated indicating that HMGA2 can be incorporated into the cellular base excision repair (BER) machinery. Increased expression of HMGA2 increased BER, and allowed cells with increased HMGA2 to be resistant to hydroxyurea, a chemotherapeutic agent for solid tumors. [22]

Interactions

HMGA2 has been shown to interact with PIAS3 [23] and NFKB1. [24]

The transport of HMGA2 to the nucleus is mediated by an interaction between its second AT-hook and importin-α2. [8]

See also

Related Research Articles

<span class="mw-page-title-main">Promoter (genetics)</span> Region of DNA encouraging transcription

In genetics, a promoter is a sequence of DNA to which proteins bind to initiate transcription of a single RNA transcript from the DNA downstream of the promoter. The RNA transcript may encode a protein (mRNA), or can have a function in and of itself, such as tRNA or rRNA. Promoters are located near the transcription start sites of genes, upstream on the DNA . Promoters can be about 100–1000 base pairs long, the sequence of which is highly dependent on the gene and product of transcription, type or class of RNA polymerase recruited to the site, and species of organism.

<span class="mw-page-title-main">CpG site</span> Region of often-methylated DNA with a cytosine followed by a guanine

The CpG sites or CG sites are regions of DNA where a cytosine nucleotide is followed by a guanine nucleotide in the linear sequence of bases along its 5' → 3' direction. CpG sites occur with high frequency in genomic regions called CpG islands.

Myc is a family of regulator genes and proto-oncogenes that code for transcription factors. The Myc family consists of three related human genes: c-myc (MYC), l-myc (MYCL), and n-myc (MYCN). c-myc was the first gene to be discovered in this family, due to homology with the viral gene v-myc.

The Let-7 microRNA precursor was identified from a study of developmental timing in C. elegans, and was later shown to be part of a much larger class of non-coding RNAs termed microRNAs. miR-98 microRNA precursor from human is a let-7 family member. Let-7 miRNAs have now been predicted or experimentally confirmed in a wide range of species (MIPF0000002). miRNAs are initially transcribed in long transcripts called primary miRNAs (pri-miRNAs), which are processed in the nucleus by Drosha and Pasha to hairpin structures of about 70 nucleotide. These precursors (pre-miRNAs) are exported to the cytoplasm by exportin5, where they are subsequently processed by the enzyme Dicer to a ~22 nucleotide mature miRNA. The involvement of Dicer in miRNA processing demonstrates a relationship with the phenomenon of RNA interference.

HMGA is a family of high mobility group proteins characterized by an AT-hook. They code for a "small, nonhistone, chromatin-associated protein that has no intrinsic transcriptional activity but can modulate transcription by altering the chromatin architecture". Mammals have two orthologs: HMGA1 and HMGA2.

<span class="mw-page-title-main">DNA-PKcs</span> Protein-coding gene in the species Homo sapiens

DNA-dependent protein kinase, catalytic subunit, also known as DNA-PKcs, is an enzyme that in humans is encoded by the gene designated as PRKDC or XRCC7. DNA-PKcs belongs to the phosphatidylinositol 3-kinase-related kinase protein family. The DNA-Pkcs protein is a serine/threonine protein kinase consisting of a single polypeptide chain of 4,128 amino acids.

<span class="mw-page-title-main">ERCC1</span> Protein-coding gene in the species Homo sapiens

DNA excision repair protein ERCC-1 is a protein that in humans is encoded by the ERCC1 gene. Together with ERCC4, ERCC1 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination.

<span class="mw-page-title-main">SOX2</span> Transcription factor gene of the SOX family

SRY -box 2, also known as SOX2, is a transcription factor that is essential for maintaining self-renewal, or pluripotency, of undifferentiated embryonic stem cells. Sox2 has a critical role in maintenance of embryonic and neural stem cells.

<span class="mw-page-title-main">RAD52</span> Protein-coding gene in the species Homo sapiens

RAD52 homolog , also known as RAD52, is a protein which in humans is encoded by the RAD52 gene.

<span class="mw-page-title-main">LPP (gene)</span> Protein-coding gene in the species Homo sapiens

Lipoma-preferred partner is a protein that in humans is encoded by the LPP gene.

<span class="mw-page-title-main">FOXN3</span> Protein-coding gene in the species Homo sapiens

Forkhead box protein N3 is a protein that in humans is encoded by the FOXN3 gene.

<span class="mw-page-title-main">LBH (gene)</span> Protein-coding gene in the species Homo sapiens

The LBH gene is a highly conserved human gene that produces the LBH protein, a transcription co-factor in the Wnt/β-catenin pathway. Upon transcriptional activation of β-catenin, LBH goes on to act as a regulator of cell proliferation and differentiation through multiple transcriptional targets. The gene is located on the p arm of chromosome 2 and is roughly 28 kb long. Current ongoing studies are examining its role in developmental and oncological settings.

<span class="mw-page-title-main">CD3EAP</span> Protein-coding gene in the species Homo sapiens

DNA-directed RNA polymerase I subunit RPA34 is an enzyme that in humans is encoded by the CD3EAP gene.

<span class="mw-page-title-main">IGF2BP2</span> Protein-coding gene in the species Homo sapiens

Insulin-like growth factor 2 mRNA-binding protein 2 is a protein that in humans is encoded by the IGF2BP2 gene.

<span class="mw-page-title-main">LHFP</span> Protein-coding gene in the species Homo sapiens

Lipoma HMGIC fusion partner is a protein that in humans is encoded by the LHFP gene.

<span class="mw-page-title-main">LHFPL1</span> Protein-coding gene in the species Homo sapiens

Lipoma HMGIC fusion partner-like 1 protein is a protein that in humans is encoded by the LHFPL1 gene.

<span class="mw-page-title-main">LHFPL2</span> Protein-coding gene in the species Homo sapiens

Lipoma HMGIC fusion partner-like 2 protein is a protein that in humans is encoded by the LHFPL2 gene.

Genome instability refers to a high frequency of mutations within the genome of a cellular lineage. These mutations can include changes in nucleic acid sequences, chromosomal rearrangements or aneuploidy. Genome instability does occur in bacteria. In multicellular organisms genome instability is central to carcinogenesis, and in humans it is also a factor in some neurodegenerative diseases such as amyotrophic lateral sclerosis or the neuromuscular disease myotonic dystrophy.

<span class="mw-page-title-main">Cancer epigenetics</span> Field of study in cancer research

Cancer epigenetics is the study of epigenetic modifications to the DNA of cancer cells that do not involve a change in the nucleotide sequence, but instead involve a change in the way the genetic code is expressed. Epigenetic mechanisms are necessary to maintain normal sequences of tissue specific gene expression and are crucial for normal development. They may be just as important, if not even more important, than genetic mutations in a cell's transformation to cancer. The disturbance of epigenetic processes in cancers, can lead to a loss of expression of genes that occurs about 10 times more frequently by transcription silencing than by mutations. As Vogelstein et al. points out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations. However, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 heavily methylated CpG islands in the promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa. Manipulation of epigenetic alterations holds great promise for cancer prevention, detection, and therapy. In different types of cancer, a variety of epigenetic mechanisms can be perturbed, such as the silencing of tumor suppressor genes and activation of oncogenes by altered CpG island methylation patterns, histone modifications, and dysregulation of DNA binding proteins. There are several medications which have epigenetic impact, that are now used in a number of these diseases.

DNA methylation in cancer plays a variety of roles, helping to change the healthy cells by regulation of gene expression to a cancer cells or a diseased cells disease pattern. One of the most widely studied DNA methylation dysregulation is the promoter hypermethylation where the CPGs islands in the promoter regions are methylated contributing or causing genes to be silenced.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000149948 - Ensembl, May 2017
  2. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  3. Ashar HR, Cherath L, Przybysz KM, Chada K (January 1996). "Genomic characterization of human HMGIC, a member of the accessory transcription factor family found at translocation breakpoints in lipomas". Genomics. 31 (2): 207–14. doi:10.1006/geno.1996.0033. PMID   8824803.
  4. Ishwad CS, Shriver MD, Lassige DM, Ferrell RE (January 1997). "The high mobility group I-C gene (HMGI-C): polymorphism and genetic localization". Human Genetics. 99 (1): 103–5. doi:10.1007/s004390050320. PMID   9003504. S2CID   42615999.
  5. 1 2 "Entrez Gene: HMGA2 high mobility group AT-hook 2".
  6. Fedele M, Battista S, Kenyon L, Baldassarre G, Fidanza V, Klein-Szanto AJ, et al. (May 2002). "Overexpression of the HMGA2 gene in transgenic mice leads to the onset of pituitary adenomas". Oncogene. 21 (20): 3190–8. doi: 10.1038/sj.onc.1205428 . PMID   12082634.
  7. 1 2 Dröge P, Davey CA (January 2008). "Do cells let-7 determine stemness?". Cell Stem Cell. 2 (1): 8–9. doi: 10.1016/j.stem.2007.12.003 . PMID   18371414.
  8. 1 2 Cattaruzzi G, Altamura S, Tessari MA, Rustighi A, Giancotti V, Pucillo C, Manfioletti G (2007). "The second AT-hook of the architectural transcription factor HMGA2 is determinant for nuclear localization and function". Nucleic Acids Research. 35 (6): 1751–60. doi:10.1093/nar/gkl1106. PMC   1874589 . PMID   17324944.
  9. Hammond SM, Sharpless NE (December 2008). "HMGA2, microRNAs, and stem cell aging". Cell. 135 (6): 1013–6. doi:10.1016/j.cell.2008.11.026. PMC   3725266 . PMID   19070572.
  10. 1 2 Mayr C, Hemann MT, Bartel DP (March 2007). "Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation". Science. 315 (5818): 1576–9. Bibcode:2007Sci...315.1576M. doi:10.1126/science.1137999. PMC   2556962 . PMID   17322030.
  11. 1 2 Fedele M, Pierantoni GM, Visone R, Fusco A (September 2006). "Critical role of the HMGA2 gene in pituitary adenomas". Cell Cycle. 5 (18): 2045–8. doi: 10.4161/cc.5.18.3211 . PMID   16969098.
  12. Meyer B, Loeschke S, Schultze A, Weigel T, Sandkamp M, Goldmann T, et al. (July 2007). "HMGA2 overexpression in non-small cell lung cancer". Molecular Carcinogenesis. 46 (7): 503–11. doi:10.1002/mc.20235. PMID   17477356. S2CID   30541611.
  13. 1 2 3 Boo LM, Lin HH, Chung V, Zhou B, Louie SG, O'Reilly MA, et al. (August 2005). "High mobility group A2 potentiates genotoxic stress in part through the modulation of basal and DNA damage-dependent phosphatidylinositol 3-kinase-related protein kinase activation". Cancer Research. 65 (15): 6622–30. doi:10.1158/0008-5472.CAN-05-0086. PMID   16061642.
  14. Shell S, Park SM, Radjabi AR, Schickel R, Kistner EO, Jewell DA, et al. (July 2007). "Let-7 expression defines two differentiation stages of cancer". Proceedings of the National Academy of Sciences of the United States of America. 104 (27): 11400–5. Bibcode:2007PNAS..10411400S. doi: 10.1073/pnas.0704372104 . PMC   2040910 . PMID   17600087.
  15. Motoyama K, Inoue H, Nakamura Y, Uetake H, Sugihara K, Mori M (April 2008). "Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family". Clinical Cancer Research. 14 (8): 2334–40. doi: 10.1158/1078-0432.CCR-07-4667 . PMID   18413822.
  16. Borrmann L, Schwanbeck R, Heyduk T, Seebeck B, Rogalla P, Bullerdiek J, Wisniewski JR (December 2003). "High mobility group A2 protein and its derivatives bind a specific region of the promoter of DNA repair gene ERCC1 and modulate its activity". Nucleic Acids Research. 31 (23): 6841–51. doi:10.1093/nar/gkg884. PMC   290254 . PMID   14627817.
  17. Facista A, Nguyen H, Lewis C, Prasad AR, Ramsey L, Zaitlin B, et al. (April 2012). "Deficient expression of DNA repair enzymes in early progression to sporadic colon cancer". Genome Integrity. 3 (1): 3. doi: 10.1186/2041-9414-3-3 . PMC   3351028 . PMID   22494821.
  18. Smith DH, Fiehn AM, Fogh L, Christensen IJ, Hansen TP, Stenvang J, et al. (March 2014). "Measuring ERCC1 protein expression in cancer specimens: validation of a novel antibody". Scientific Reports. 4: 4313. Bibcode:2014NatSR...4E4313S. doi:10.1038/srep04313. PMC   3945488 . PMID   24603753.
  19. Prasad AR, Prasad S, Nguyen H, Facista A, Lewis C, Zaitlin B, et al. (July 2014). "Novel diet-related mouse model of colon cancer parallels human colon cancer". World Journal of Gastrointestinal Oncology. 6 (7): 225–43. doi: 10.4251/wjgo.v6.i7.225 . PMC   4092339 . PMID   25024814.
  20. Zha L, Wang Z, Tang W, Zhang N, Liao G, Huang Z (May 2012). "Genome-wide analysis of HMGA2 transcription factor binding sites by ChIP on chip in gastric carcinoma cells". Molecular and Cellular Biochemistry. 364 (1–2): 243–51. doi:10.1007/s11010-012-1224-z. PMID   22246783. S2CID   15777147.
  21. Li AY, Boo LM, Wang SY, Lin HH, Wang CC, Yen Y, et al. (July 2009). "Suppression of nonhomologous end joining repair by overexpression of HMGA2". Cancer Research. 69 (14): 5699–706. doi:10.1158/0008-5472.CAN-08-4833. PMC   2737594 . PMID   19549901.
  22. Summer H, Li O, Bao Q, Zhan L, Peter S, Sathiyanathan P, et al. (July 2009). "HMGA2 exhibits dRP/AP site cleavage activity and protects cancer cells from DNA-damage-induced cytotoxicity during chemotherapy". Nucleic Acids Research. 37 (13): 4371–84. doi:10.1093/nar/gkp375. PMC   2715238 . PMID   19465398.
  23. Zentner MD, Lin HH, Deng HT, Kim KJ, Shih HM, Ann DK (August 2001). "Requirement for high mobility group protein HMGI-C interaction with STAT3 inhibitor PIAS3 in repression of alpha-subunit of epithelial Na+ channel (alpha-ENaC) transcription by Ras activation in salivary epithelial cells". The Journal of Biological Chemistry. 276 (32): 29805–14. doi: 10.1074/jbc.M103153200 . PMID   11390395.
  24. Noro B, Licheri B, Sgarra R, Rustighi A, Tessari MA, Chau KY, et al. (April 2003). "Molecular dissection of the architectural transcription factor HMGA2". Biochemistry. 42 (15): 4569–77. doi:10.1021/bi026605k. PMID   12693954. S2CID   39605320.

Further reading

This article incorporates text from the United States National Library of Medicine, which is in the public domain.