Jump to content
 







Main menu
   


Navigation  



Main page
Contents
Current events
Random article
About Wikipedia
Contact us
Donate
 




Contribute  



Help
Learn to edit
Community portal
Recent changes
Upload file
 








Search  

































Create account

Log in
 









Create account
 Log in
 




Pages for logged out editors learn more  



Contributions
Talk
 



















Contents

   



(Top)
 


1 Function  





2 Regulation by let-7  





3 Clinical significance  



3.1  Relationship with cancer  





3.2  ERCC1  





3.3  Chromatin immunoprecipitation  





3.4  Non-homologous end joining DNA repair  





3.5  Base excision repair pathway  







4 Interactions  





5 See also  





6 References  





7 Further reading  





8 External links  














HMGA2






Galego
Татарча / tatarça
Українська
 

Edit links
 









Article
Talk
 

















Read
Edit
View history
 








Tools
   


Actions  



Read
Edit
View history
 




General  



What links here
Related changes
Upload file
Special pages
Permanent link
Page information
Cite this page
Get shortened URL
Download QR code
Wikidata item
 




Print/export  



Download as PDF
Printable version
 
















Appearance
   

 






From Wikipedia, the free encyclopedia
 


HMGA2

Identifiers

Aliases

HMGA2, BABL, HMGI-C, HMGIC, LIPO, STQTL9, high mobility group AT-hook 2, SRS5

External IDs

OMIM: 600698; MGI: 101761; HomoloGene: 136767; GeneCards: HMGA2; OMA:HMGA2 - orthologs

Gene location (Human)

Chromosome 12 (human)

Chr.

Chromosome 12 (human)[1]

Chromosome 12 (human)

Genomic location for HMGA2

Genomic location for HMGA2

Band

12q14.3

Start

65,824,460 bp[1]

End

65,966,291 bp[1]

Gene location (Mouse)

Chromosome 10 (mouse)

Chr.

Chromosome 10 (mouse)[2]

Chromosome 10 (mouse)

Genomic location for HMGA2

Genomic location for HMGA2

Band

10 D2|10 67.94 cM

Start

120,197,180 bp[2]

End

120,312,374 bp[2]

Bgee

Mouse (ortholog)


  • embryo

  • stromal cell of endometrium

  • ventricular zone

  • cartilage tissue

  • ganglionic eminence

  • testicle

  • retinal pigment epithelium

  • buccal mucosa cell

  • amniotic fluid

  • endothelial cell of lymphatic vessel

  • tail of embryo

  • abdominal wall

  • maxillary prominence

  • mandibular prominence

  • primitive streak

  • somite

  • hand

  • medullary collecting duct
  • More reference expression data

    BioGPS

    n/a

    Molecular function

  • DNA-dependent protein kinase activity
  • MH1 domain binding
  • 5'-deoxyribose-5-phosphate lyase activity
  • minor groove of adenine-thymine-rich DNA binding
  • DNA-binding transcription activator activity, RNA polymerase II-specific
  • transcription factor binding
  • C2H2 zinc finger domain binding
  • DNA-binding transcription repressor activity, RNA polymerase II-specific
  • MH2 domain binding
  • protein binding
  • DNA-(apurinic or apyrimidinic site) endonuclease activity
  • nucleosomal DNA binding
  • DNA binding, bending
  • SMAD binding
  • cAMP response element binding
  • RNA polymerase II cis-regulatory region sequence-specific DNA binding
  • DNA-binding transcription factor activity, RNA polymerase II-specific
  • transcription coregulator activity
  • Cellular component

  • senescence-associated heterochromatin focus
  • nuclear chromosome
  • protein-DNA complex
  • chromatin
  • nucleus
  • nucleoplasm
  • Biological process

  • positive regulation of transcription regulatory region DNA binding
  • regulation of cell cycle process
  • regulation of transcription, DNA-templated
  • endodermal cell differentiation
  • chondrocyte differentiation
  • regulation of cellular response to drug
  • negative regulation of DNA binding
  • response to virus
  • regulation of stem cell population maintenance
  • negative regulation of apoptotic process
  • positive regulation of cellular response to X-ray
  • negative regulation of transcription by RNA polymerase II
  • chromatin organization
  • heterochromatin assembly
  • oncogene-induced cell senescence
  • stem cell differentiation
  • transcription, DNA-templated
  • multicellular organism development
  • positive regulation of transcription, DNA-templated
  • negative regulation by host of viral transcription
  • cell division
  • positive regulation of gene expression
  • mesenchymal cell differentiation
  • chromosome condensation
  • negative regulation of double-strand break repair via nonhomologous end joining
  • chondrocyte proliferation
  • positive regulation of cellular senescence
  • positive regulation of response to DNA damage stimulus
  • epithelial to mesenchymal transition
  • positive regulation of apoptotic process
  • mesodermal cell differentiation
  • regulation of growth
  • cell cycle
  • mitotic G2 DNA damage checkpoint signaling
  • negative regulation of transcription, DNA-templated
  • positive regulation of stem cell proliferation
  • fat cell differentiation
  • chromosome breakage
  • mesodermal-endodermal cell signaling
  • base-excision repair
  • negative regulation of single stranded viral RNA replication via double stranded DNA intermediate
  • transcription by RNA polymerase II
  • positive regulation of angiogenesis
  • positive regulation of cell proliferation in bone marrow
  • negative regulation of cellular senescence
  • positive regulation of protein serine/threonine kinase activity
  • Sources:Amigo / QuickGO

    Species

    Human

    Mouse

    Entrez

    Ensembl

    UniProt

    RefSeq (mRNA)

    NM_001330190

    NM_010441
    NM_001347170

    RefSeq (protein)

    NP_001287847
    NP_001287848
    NP_001317119
    NP_003474
    NP_003475

    NP_001334099
    NP_034571

    Location (UCSC)

    Chr 12: 65.82 – 65.97 Mb

    Chr 10: 120.2 – 120.31 Mb

    PubMed search

    [3]

    [4]

    Wikidata

    High-mobility group AT-hook 2, also known as HMGA2, is a protein that, in humans, is encoded by the HMGA2 gene.[5][6][7]

    Function[edit]

    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.[7]

    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.[8] The microRNA let-7 is largely responsible for this time-dependent regulation of HMGA2.[9] 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),[10] and genome-wide association studies linking HMGA2-associated SNPs to variation in human height.[11]

    Regulation by let-7[edit]

    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).[12] 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.[9]

    Clinical significance[edit]

    Relationship with cancer[edit]

    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.[13][14] The same mutations that lead to pituitary adenomas in mice can be found in similar cancers in humans.[13] Its presence is associated with poor prognosis for the patient, but also with sensitization of the cancer cells to certain forms of cancer therapy.[15] 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.[15] However, the expression of HMGA2 is also associated with increased rates of metastasisinbreast 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.[15]

    A very common finding in HMGA2-high cancers is the under-expression of let-7.[16] 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.[12]

    ERCC1[edit]

    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.[17] (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.,[18] 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.[19][20] ERCC1 protein expression was also reduced in a diet-related mouse model of colon cancer.[21] 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[edit]

    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.[22] 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[edit]

    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.[23] showed that

    Base excision repair pathway[edit]

    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.[24]

    Interactions[edit]

    HMGA2 has been shown to interact with PIAS3[25] and NFKB1.[26]

    The transport of HMGA2 to the nucleus is mediated by an interaction between its second AT-hook and importin-α2.[10]

    See also[edit]

    References[edit]

  • ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000056758Ensembl, May 2017
  • ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  • ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  • ^ 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.
  • ^ 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.
  • ^ a b "Entrez Gene: HMGA2 high mobility group AT-hook 2".
  • ^ 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.
  • ^ a b 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.
  • ^ a b 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.
  • ^ 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.
  • ^ a b 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.
  • ^ a b 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.
  • ^ 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.
  • ^ a b c 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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.
  • ^ 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[edit]

    • Pedeutour F, Ligon AH, Morton CC (November 1999). "[Genetics of uterine leiomyomata]". Bulletin du Cancer. 86 (11): 920–8. PMID 10586108.
  • Reeves R, Beckerbauer L (May 2001). "HMGI/Y proteins: flexible regulators of transcription and chromatin structure". Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression. 1519 (1–2): 13–29. doi:10.1016/S0167-4781(01)00215-9. PMID 11406267.
  • Manfioletti G, Giancotti V, Bandiera A, Buratti E, Sautière P, Cary P, et al. (December 1991). "cDNA cloning of the HMGI-C phosphoprotein, a nuclear protein associated with neoplastic and undifferentiated phenotypes". Nucleic Acids Research. 19 (24): 6793–7. doi:10.1093/nar/19.24.6793. PMC 329311. PMID 1762909.
  • Chau KY, Patel UA, Lee KL, Lam HY, Crane-Robinson C (November 1995). "The gene for the human architectural transcription factor HMGI-C consists of five exons each coding for a distinct functional element". Nucleic Acids Research. 23 (21): 4262–6. doi:10.1093/nar/23.21.4262. PMC 307378. PMID 7501444.
  • Schoenmakers EF, Mols R, Wanschura S, Kools PF, Geurts JM, Bartnitzke S, et al. (October 1994). "Identification, molecular cloning, and characterization of the chromosome 12 breakpoint cluster region of uterine leiomyomas". Genes, Chromosomes & Cancer. 11 (2): 106–18. doi:10.1002/gcc.2870110207. PMID 7529547. S2CID 25942810.
  • Ashar HR, Fejzo MS, Tkachenko A, Zhou X, Fletcher JA, Weremowicz S, et al. (July 1995). "Disruption of the architectural factor HMGI-C: DNA-binding AT hook motifs fused in lipomas to distinct transcriptional regulatory domains". Cell. 82 (1): 57–65. doi:10.1016/0092-8674(95)90052-7. PMID 7606786. S2CID 15593143.
  • Schoenmakers EF, Wanschura S, Mols R, Bullerdiek J, Van den Berghe H, Van de Ven WJ (August 1995). "Recurrent rearrangements in the high mobility group protein gene, HMGI-C, in benign mesenchymal tumours". Nature Genetics. 10 (4): 436–44. doi:10.1038/ng0895-436. PMID 7670494. S2CID 29935721.
  • Patel UA, Bandiera A, Manfioletti G, Giancotti V, Chau KY, Crane-Robinson C (May 1994). "Expression and cDNA cloning of human HMGI-C phosphoprotein". Biochemical and Biophysical Research Communications. 201 (1): 63–70. doi:10.1006/bbrc.1994.1669. PMID 8198613.
  • 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.
  • 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.
  • Petit MM, Swarts S, Bridge JA, Van de Ven WJ (October 1998). "Expression of reciprocal fusion transcripts of the HMGIC and LPP genes in parosteal lipoma". Cancer Genetics and Cytogenetics. 106 (1): 18–23. doi:10.1016/S0165-4608(98)00038-7. PMID 9772904.
  • Schoenmakers EF, Huysmans C, Van de Ven WJ (January 1999). "Allelic knockout of novel splice variants of human recombination repair gene RAD51B in t(12;14) uterine leiomyomas". Cancer Research. 59 (1): 19–23. PMID 9892177.
  • Gattas GJ, Quade BJ, Nowak RA, Morton CC (August 1999). "HMGIC expression in human adult and fetal tissues and in uterine leiomyomata". Genes, Chromosomes & Cancer. 25 (4): 316–22. doi:10.1002/(SICI)1098-2264(199908)25:4<316::AID-GCC2>3.0.CO;2-0. PMID 10398424. S2CID 42957480.
  • Schwanbeck R, Manfioletti G, Wiśniewski JR (January 2000). "Architecture of high mobility group protein I-C.DNA complex and its perturbation upon phosphorylation by Cdc2 kinase". The Journal of Biological Chemistry. 275 (3): 1793–801. doi:10.1074/jbc.275.3.1793. PMID 10636877.
  • Piekielko A, Drung A, Rogalla P, Schwanbeck R, Heyduk T, Gerharz M, et al. (January 2001). "Distinct organization of DNA complexes of various HMGI/Y family proteins and their modulation upon mitotic phosphorylation". The Journal of Biological Chemistry. 276 (3): 1984–92. doi:10.1074/jbc.M004065200. PMID 11034995.
  • Rogalla P, Lemke I, Kazmierczak B, Bullerdiek J (December 2000). "An identical HMGIC-LPP fusion transcript is consistently expressed in pulmonary chondroid hamartomas with t(3;12)(q27-28;q14-15)". Genes, Chromosomes & Cancer. 29 (4): 363–6. doi:10.1002/1098-2264(2000)9999:9999<1::AID-GCC1043>3.0.CO;2-N. PMID 11066083. S2CID 35883889.
  • 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.
  • Röijer E, Nordkvist A, Ström AK, Ryd W, Behrendt M, Bullerdiek J, et al. (February 2002). "Translocation, deletion/amplification, and expression of HMGIC and MDM2 in a carcinoma ex pleomorphic adenoma". The American Journal of Pathology. 160 (2): 433–40. doi:10.1016/S0002-9440(10)64862-6. PMC 1850659. PMID 11839563.
  • External links[edit]

    (1) Basic domains

    (1.1) Basic leucine zipper (bZIP)

  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • AP-1
  • BACH
  • BATF
  • BLZF1
  • C/EBP
  • CREB
  • CREM
  • DBP
  • DDIT3
  • GABPA
  • GCN4
  • HLF
  • MAF
  • NFE
  • NFIL3
  • NRL
  • NRF
  • XBP1
  • (1.2) Basic helix-loop-helix (bHLH)

    Group A

  • ASCL2
  • ATOH1
  • HAND
  • MESP2
  • Myogenic regulatory factors
  • NeuroD
  • Neurogenins
  • OLIG
  • Paraxis
  • SLC
  • Twist
  • Group B

  • Myc
  • MXD4
  • TCF4
  • Group C
    bHLH-PAS

  • AHRR
  • ARNT
  • CLOCK
  • HIF
  • NPAS
  • PER
  • SIM
  • Group D

    • BHLH
  • 3
  • 9
  • Pho4
  • ID
  • Group E

    • HES
  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • HEY
  • Group F
    bHLH-COE

    (1.3) bHLH-ZIP

  • MAX
  • MITF
  • MNT
  • MLX
  • MLXIPL
  • MXI1
  • Myc
  • SREBP
  • USF1
  • (1.4) NF-1

  • B
  • C
  • X
  • SMAD
  • (1.5) RF-X

    • RFX
  • 2
  • 3
  • 4
  • 5
  • 6
  • ANK
  • (1.6) Basic helix-span-helix (bHSH)

  • β
  • γ
  • δ
  • ε
  • (2) Zinc finger DNA-binding domains

    (2.1) Nuclear receptor (Cys4)

    subfamily 1

  • β
  • CAR
  • FXR
  • LXR
  • PPAR
  • PXR
  • RAR
  • ROR
  • Rev-ErbA
  • VDR
  • subfamily 2

  • II
  • Ear-2
  • HNF4
  • PNR
  • RXR
  • Testicular receptor
  • TLX
  • subfamily 3

  • Estrogen
  • Glucocorticoid
  • Mineralocorticoid
  • Progesterone
  • Estrogen related
  • subfamily 4

  • NOR1
  • NURR1
  • subfamily 5

  • SF1
  • subfamily 6

    subfamily 0

  • SHP
  • (2.2) Other Cys4

  • 2
  • 3
  • 4
  • 5
  • 6
  • MTA
  • TRPS1
  • (2.3) Cys2His2

  • TFIIB
  • TFIID
  • TFIIE
  • TFIIF
  • 1
  • 2
  • (2.4) Cys6

    (2.5) Alternating composition

  • DIDO1
  • GRLF1
  • ING
  • JARID
  • JMJD1B
  • (2.6) WRKY

    (3) Helix-turn-helix domains

    Antennapedia
    ANTP class

    protoHOX
    Hox-like

  • 2
  • Xlox
  • Cdx
  • 2
  • 4
  • extended Hox: Evx1
  • Evx2
  • MEOX1
  • MEOX2
  • Homeobox
  • GBX1
  • GBX2
  • MNX1
  • metaHOX
    NK-like

  • BARHL2
  • BARX1
  • BARX2
  • BSX
  • DBX
  • DLX
  • EMX
  • EN
  • HHEX
  • HLX
  • LBX1
  • LBX2
  • MSX
  • NANOG
  • NKX
  • NATO
  • TLX1
  • TLX2
  • TLX3
  • VAX1
  • VAX2
  • other

  • CRX
  • CUTL1
  • FHL
  • HESX1
  • HOPX
  • LMX
  • NOBOX
  • TALE
  • PHF
  • POU domain
  • SATB2
  • ZEB
  • (3.2) Paired box

  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • 8
  • 9
  • PRRX
  • PROP1
  • PHOX
  • RAX
  • SHOX
  • SHOX2
  • VSX1
  • VSX2
  • Bicoid
  • (3.3) Fork head / winged helix

  • 2
  • 3
  • 4
  • 5
  • FOX proteins
  • (3.4) Heat shock factors

  • 2
  • 4
  • (3.5) Tryptophan clusters

    • ELF
  • 4
  • 5
  • EGF
  • ELK
  • ERF
  • ETS
  • ETV
  • FLI1
  • Interferon regulatory factors
  • MYB
  • MYBL2
  • (3.6) TEA domain

    • transcriptional enhancer factor
  • 2
  • 3
  • 4
  • (4) β-Scaffold factors with minor groove contacts

  • NFKB2
  • REL
  • RELA
  • RELB
  • NFAT
  • (4.2) STAT

  • 2
  • 3
  • 4
  • 5
  • 6
  • (4.3) p53-like

  • TP63
  • p73
  • TBX
  • MYRF
  • (4.4) MADS box

  • B
  • C
  • D
  • SRF
  • (4.6) TATA-binding proteins

  • TBPL1
  • (4.7) High-mobility group

  • HMGB
  • HMGN
  • HNF
  • SOX
  • SRY
  • SSRP1
  • TCF/LEF
  • TOX
  • (4.9) Grainyhead

    (4.10) Cold-shock domain

  • YBX1
  • (4.11) Runt

  • CBFA2T3
  • RUNX1
  • RUNX2
  • RUNX3
  • RUNX1T1
  • (0) Other transcription factors

    (0.2) HMGI(Y)

  • 2
  • HBP1
  • (0.3) Pocket domain

  • RBL1
  • RBL2
  • (0.5) AP-2/EREBP-related factors

  • EREBP
  • B3
  • (0.6) Miscellaneous

  • 1B
  • 2
  • 3A
  • 3B
  • 4A
  • CAP
  • IFI
  • MLL
  • MNDA
  • NFY
  • Rho/Sigma
  • see also transcription factor/coregulator deficiencies

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


    Retrieved from "https://en.wikipedia.org/w/index.php?title=HMGA2&oldid=1174463551"

    Categories: 
    Genes on human chromosome 12
    Transcription factors
    Hidden categories: 
    Articles with short description
    Short description matches Wikidata
    Wikipedia articles incorporating text from the United States National Library of Medicine
    Use dmy dates from April 2017
     



    This page was last edited on 8 September 2023, at 16:03 (UTC).

    Text is available under the Creative Commons Attribution-ShareAlike License 4.0; additional terms may apply. By using this site, you agree to the Terms of Use and Privacy Policy. Wikipedia® is a registered trademark of the Wikimedia Foundation, Inc., a non-profit organization.



    Privacy policy

    About Wikipedia

    Disclaimers

    Contact Wikipedia

    Code of Conduct

    Developers

    Statistics

    Cookie statement

    Mobile view



    Wikimedia Foundation
    Powered by MediaWiki