AGGF1: Difference between revisions

Jump to navigation Jump to search
m (Disambiguated: TranscriptionTranscription (biology))
imported>Citation bot
m (Alter: journal. Add: pmc. You can use this bot yourself. Report bugs here. | Headbomb)
 
Line 1: Line 1:
{{Infobox_gene|Gene ontology=cell adhesion
{{Infobox gene|Gene ontology=cell adhesion
positive regulation of angiogenesis
positive regulation of angiogenesis
endothelial cell proliferation}}
endothelial cell proliferation}}
'''Angiogenic factor with G patch and FHA domains 1''' is a [[protein]] that in humans is encoded by the ''AGGF1'' [[gene]].<ref name="pmid18564129">{{cite journal | vauthors = Hu Y, Li L, Seidelmann SB, Timur AA, Shen PH, Driscoll DJ, Wang QK | title = Identification of association of common AGGF1 variants with susceptibility for Klippel-Trenaunay syndrome using the structure association program | journal = Annals of Human Genetics | volume = 72 | issue = Pt 5 | pages = 636–43 | date = September 2008 | pmid = 18564129 | pmc = 2602961 | doi = 10.1111/j.1469-1809.2008.00458.x }}</ref><ref name="pmid17103452">{{cite journal | vauthors = Gutierrez S, Magano L, Delicado A, Mori MA, de Torres ML, Fernández L, Palomares M, Fernández E, Tarduchy GR, Molano J, Gracia R, Pajares IL, Lapunzina P | title = The G397A (E133K) change in the AGGF1 (VG5Q) gene is a single nucleotide polymorphism in the Spanish population | journal = American Journal of Medical Genetics. Part A | volume = 140 | issue = 24 | pages = 2832–3 | date = December 2006 | pmid = 17103452 | doi = 10.1002/ajmg.a.31532 }}</ref><ref name="entrez">{{cite web | title = Entrez Gene: AGGF1 angiogenic factor with G patch and FHA domains 1| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55109| accessdate = }}</ref>
'''Angiogenic factor with G patch and FHA domains 1''' is a [[protein]] that in humans is encoded by the ''AGGF1'' [[gene]].<ref name="pmid18564129">{{cite journal | vauthors = Hu Y, Li L, Seidelmann SB, Timur AA, Shen PH, Driscoll DJ, Wang QK | title = Identification of association of common AGGF1 variants with susceptibility for Klippel-Trenaunay syndrome using the structure association program | journal = Annals of Human Genetics | volume = 72 | issue = Pt 5 | pages = 636–43 | date = September 2008 | pmid = 18564129 | pmc = 2602961 | doi = 10.1111/j.1469-1809.2008.00458.x }}</ref><ref name="pmid17103452">{{cite journal | vauthors = Gutierrez S, Magano L, Delicado A, Mori MA, de Torres ML, Fernández L, Palomares M, Fernández E, Tarduchy GR, Molano J, Gracia R, Pajares IL, Lapunzina P | title = The G397A (E133K) change in the AGGF1 (VG5Q) gene is a single nucleotide polymorphism in the Spanish population | journal = American Journal of Medical Genetics. Part A | volume = 140 | issue = 24 | pages = 2832–3 | date = December 2006 | pmid = 17103452 | doi = 10.1002/ajmg.a.31532 }}</ref><ref name="entrez">{{cite web | title = Entrez Gene: AGGF1 angiogenic factor with G patch and FHA domains 1| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55109| accessdate = }}</ref>


AGGF1 is a human gene that functions as an [[Angiogenesis|angiogenic]] factor with a G-patch and [[forkhead-associated domain]].<ref>{{cite journal | vauthors = Zhan M, Hori Y, Wada N, Ikeda J, Hata Y, Osuga K, Morii E | title = Angiogenic Factor with G-patch and FHA Domain 1 (AGGF1) Expression in Human Vascular Lesions | journal = Acta Histochemica Et Cytochemica | volume = 49 | issue = 2 | pages = 75–81 | date = April 2016 | pmid = 27222614 | pmc = 4858542 | doi = 10.1267/ahc.15035 }}</ref> This gene is predominantly expressed in activated, plump [[endothelial cells]] and acts to regulate [[angiogenesis]] and vascular development.<ref name="Zhang_2016">{{cite journal | vauthors = Zhang T, Yao Y, Wang J, Li Y, He P, Pasupuleti V, Hu Z, Jia X, Song Q, Tian XL, Hu C, Chen Q, Wang QK | title = Haploinsufficiency of Klippel-Trenaunay syndrome gene Aggf1 inhibits developmental and pathological angiogenesis by inactivating PI3K and AKT and disrupts vascular integrity by activating VE-cadherin | journal = Human Molecular Genetics | volume = 25 | issue = 23 | pages = 5094–5110 | date = December 2016 | pmid = 27522498 | doi = 10.1093/hmg/ddw273 }}</ref> AGGF1 is known to interact with a wide range of proteins involved in vascular development.<ref name="Li_2014">{{cite journal | vauthors = Li L, Chen D, Li J, Wang X, Wang N, Xu C, Wang QK | title = Aggf1 acts at the top of the genetic regulatory hierarchy in specification of hemangioblasts in zebrafish | journal = Blood | volume = 123 | issue = 4 | pages = 501–8 | date = January 2014 | pmid = 24277077 | pmc = 3901065 | doi = 10.1182/blood-2013-07-514612 }}</ref> Mutations to AGGF1 have been implicated in multiple cancers and is known to cause the rare congenital condition, [[Klippel–Trénaunay syndrome|Klippel-Trenaunay syndrome]].<ref name="Zhang_2016" /><ref name="Yao_2017">{{cite journal | vauthors = Yao HH, Wang BJ, Wu Y, Huang Q | title = High Expression of Angiogenic Factor with G-Patch and FHA Domain1 (AGGF1) Predicts Poor Prognosis in Gastric Cancer | journal = Medical Science Monitor : International Medical Journal of Experimental and Clinical Research | volume = 23 | issue = | pages = 1286–1294 | year = 2017 | pmid = 28289272 | pmc = 5362190 }}</ref><ref name="Wang_2015">{{cite journal | vauthors = Wang W, Li GY, Zhu JY, Huang DB, Zhou HC, Zhong W, Ji CS | title = Overexpression of AGGF1 is correlated with angiogenesis and poor prognosis of hepatocellular carcinoma | journal = Medical Oncology | volume = 32 | issue = 4 | pages = 131 | date = April 2015 | pmid = 25796501 | doi = 10.1007/s12032-015-0574-2 }}</ref>
AGGF1 is a human gene that functions as an [[Angiogenesis|angiogenic]] factor with a G-patch and [[forkhead-associated domain]].<ref name="Zhan_2016"/> This gene is predominantly expressed in activated, plump [[endothelial cells]] and acts to regulate [[angiogenesis]] and vascular development.<ref name="Zhang_2016">{{cite journal | vauthors = Zhang T, Yao Y, Wang J, Li Y, He P, Pasupuleti V, Hu Z, Jia X, Song Q, Tian XL, Hu C, Chen Q, Wang QK | title = Haploinsufficiency of Klippel-Trenaunay syndrome gene Aggf1 inhibits developmental and pathological angiogenesis by inactivating PI3K and AKT and disrupts vascular integrity by activating VE-cadherin | journal = Human Molecular Genetics | volume = 25 | issue = 23 | pages = 5094–5110 | date = December 2016 | pmid = 27522498 | pmc = 6078640 | doi = 10.1093/hmg/ddw273 }}</ref> AGGF1 is known to interact with a wide range of proteins involved in vascular development.<ref name="Li_2014">{{cite journal | vauthors = Li L, Chen D, Li J, Wang X, Wang N, Xu C, Wang QK | title = Aggf1 acts at the top of the genetic regulatory hierarchy in specification of hemangioblasts in zebrafish | journal = Blood | volume = 123 | issue = 4 | pages = 501–8 | date = January 2014 | pmid = 24277077 | pmc = 3901065 | doi = 10.1182/blood-2013-07-514612 }}</ref> Mutations to AGGF1 have been implicated in multiple cancers and is known to cause the rare congenital condition, [[Klippel–Trénaunay syndrome|Klippel-Trenaunay syndrome]].<ref name="Zhang_2016" /><ref name="Yao_2017">{{cite journal | vauthors = Yao HH, Wang BJ, Wu Y, Huang Q | title = High Expression of Angiogenic Factor with G-Patch and FHA Domain1 (AGGF1) Predicts Poor Prognosis in Gastric Cancer | journal = Medical Science Monitor : International Medical Journal of Experimental and Clinical Research | volume = 23 | issue = | pages = 1286–1294 | year = 2017 | pmid = 28289272 | pmc = 5362190 | doi=10.12659/msm.903248}}</ref><ref name="Wang_2015">{{cite journal | vauthors = Wang W, Li GY, Zhu JY, Huang DB, Zhou HC, Zhong W, Ji CS | title = Overexpression of AGGF1 is correlated with angiogenesis and poor prognosis of hepatocellular carcinoma | journal = Medical Oncology | volume = 32 | issue = 4 | pages = 131 | date = April 2015 | pmid = 25796501 | doi = 10.1007/s12032-015-0574-2 }}</ref>


== Gene ==
== Gene ==
The gene was originally named VG5Q, indicating that it was a vascular gene on chromosome 5, but the name was later changed to reflect its function, instead of just its location.<ref name="Fan_2009" />
The gene was originally named VG5Q, indicating that it was a vascular gene on chromosome 5, but the name was later changed to reflect its function, instead of just its location.<ref name="Fan_2009" />


The AGGF1 gene promoter does not contain [[TATA box]] and contains 2 [[Transcription (biology)|transcription]] start sites that are -367 and -364 base pairs ahead of the base translation start site.<ref name="Fan_2009">{{cite journal | vauthors = Fan C, Ouyang P, Timur AA, He P, You SA, Hu Y, Ke T, Driscoll DJ, Chen Q, Wang QK | title = Novel roles of GATA1 in regulation of angiogenic factor AGGF1 and endothelial cell function | journal = The Journal of Biological Chemistry | volume = 284 | issue = 35 | pages = 23331–43 | date = August 2009 | pmid = 19556247 | pmc = 2749107 | doi = 10.1074/jbc.M109.036079 }}</ref> The gene promoter contains over 50 [[CpG site|CpG islands]], which makes it [[DNA methylation]] target.<ref name="Fan_2009" /> AGGF1 is regulated by 2 repressor sites and 2 activator sites.<ref name="Fan_2009" /> While the presence of 2 repressor and 2 activator sites is clear, the only known transcription factor that regulates AGGF1 is GATA1.<ref name="Fan_2009" /> GATA1 binds upstream of the AGGF1 gene promoter at -295 and -300, and the binding of GATA1 will lead to increased AGGF1 expression.<ref name="Zhang_2016" /><ref name="Fan_2009" /> For the gene to be fully expressed, both of the activator sites must be bound by the transcription factors, GATA1 and another unknown factor.<ref name="Fan_2009" />
The AGGF1 gene promoter does not contain a [[TATA box]] and contains 2 [[Transcription (biology)|transcription]] start sites that are -367 and -364 base pairs ahead of the base translation start site.<ref name="Fan_2009">{{cite journal | vauthors = Fan C, Ouyang P, Timur AA, He P, You SA, Hu Y, Ke T, Driscoll DJ, Chen Q, Wang QK | title = Novel roles of GATA1 in regulation of angiogenic factor AGGF1 and endothelial cell function | journal = The Journal of Biological Chemistry | volume = 284 | issue = 35 | pages = 23331–43 | date = August 2009 | pmid = 19556247 | pmc = 2749107 | doi = 10.1074/jbc.M109.036079 }}</ref> The gene promoter contains over 50 [[CpG site|CpG islands]], which makes it a [[DNA methylation]] target.<ref name="Fan_2009" /> AGGF1 is regulated by 2 repressor sites and 2 activator sites.<ref name="Fan_2009" /> While the presence of 2 repressor and 2 activator sites is clear, the only known transcription factor that regulates AGGF1 is GATA1.<ref name="Fan_2009" /> GATA1 binds upstream of the AGGF1 gene promoter at -295 and -300, and the binding of GATA1 will lead to increased AGGF1 expression.<ref name="Zhang_2016" /><ref name="Fan_2009" /> For the gene to be fully expressed, both of the activator sites must be bound by the transcription factors, GATA1 and another unknown factor.<ref name="Fan_2009" />
 
== Protein ==
== Protein ==
To form a protein, an mRNA transcript must be [[Transcription (biology)|transcribed]] from the DNA. For AGGF1, the mRNA transcript contains 14 exons and 34 807 nucleotides.<ref name="Hu_2008">{{cite journal | vauthors = Hu Y, Li L, Seidelmann SB, Timur AA, Shen PH, Driscoll DJ, Wang QK | title = Identification of association of common AGGF1 variants with susceptibility for Klippel-Trenaunay syndrome using the structure association program | journal = Annals of Human Genetics | volume = 72 | issue = Pt 5 | pages = 636–43 | date = September 2008 | pmid = 18564129 | pmc = 2602961 | doi = 10.1111/j.1469-1809.2008.00458.x }}</ref>
To form a protein, an mRNA transcript must be [[Transcription (biology)|transcribed]] from the DNA. For AGGF1, the mRNA transcript contains 14 exons and 34 807 nucleotides.<ref name="pmid18564129"/>


There are 714 amino acids present in this protein, and it has a molecular weight of 80997 Da.<ref name="Liu_2014" /> It contains [[Coiled coil|coiled coil domain]] at positions 18-88 and an OCRE domain at the [[N-terminus|N terminus]].<ref name="Liu_2014">{{cite journal | vauthors = Liu Y, Yang H, Song L, Li N, Han QY, Tian C, Gao E, Du J, Xia YL, Li HH | title = AGGF1 protects from myocardial ischemia/reperfusion injury by regulating myocardial apoptosis and angiogenesis | journal = Apoptosis | volume = 19 | issue = 8 | pages = 1254–68 | date = August 2014 | pmid = 24893993 | doi = 10.1007/s10495-014-1001-4 }}</ref>  The G-patch domain is located at amino acids 619-663 while the forkhead-associated domain is located at amino acids 435-508.<ref name="Liu_2014" /> While it is known that these domains are present in the protein, their role in protein function remains unclear.
There are 714 amino acids present in this protein, and it has a molecular weight of 80997 Da.<ref name="Liu_2014" /> It contains a [[Coiled coil|coiled coil domain]] at positions 18-88 and an OCRE domain at the [[N-terminus|N terminus]].<ref name="Liu_2014">{{cite journal | vauthors = Liu Y, Yang H, Song L, Li N, Han QY, Tian C, Gao E, Du J, Xia YL, Li HH | title = AGGF1 protects from myocardial ischemia/reperfusion injury by regulating myocardial apoptosis and angiogenesis | journal = Apoptosis | volume = 19 | issue = 8 | pages = 1254–68 | date = August 2014 | pmid = 24893993 | doi = 10.1007/s10495-014-1001-4 }}</ref>  The G-patch domain is located at amino acids 619-663 while the forkhead-associated domain is located at amino acids 435-508.<ref name="Liu_2014" /> While it is known that these domains are present in the protein, their role in protein function remains unclear.


AGGF1 was the third [[Haploinsufficiency|haploinsufficient]] human gene identified.<ref name="Zhang_2016" /> Haploinsufficiency means AGGF1 is "dose dependent" so any reductions in protein product can have [[Phenotypic trait|phenotypic]] consequences on the vascular development of the organism.  
AGGF1 was the third [[Haploinsufficiency|haploinsufficient]] human gene identified.<ref name="Zhang_2016" /> Haploinsufficiency means AGGF1 is "dose dependent" so any reductions in protein product can have [[Phenotypic trait|phenotypic]] consequences on the vascular development of the organism.


== Expression ==
== Expression ==
AGGF1 is largely expressed during early embryonic vein specification, and expression is increased when endothelial cells are activated.<ref name="Liu_2014" /><ref name="Zhan_2016">{{cite journal | vauthors = Zhan M, Hori Y, Wada N, Ikeda J, Hata Y, Osuga K, Morii E | title = Angiogenic Factor with G-patch and FHA Domain 1 (AGGF1) Expression in Human Vascular Lesions | journal = Acta Histochemica Et Cytochemica | volume = 49 | issue = 2 | pages = 75–81 | date = April 2016 | pmid = 27222614 | pmc = 4858542 | doi = 10.1267/ahc.15035 }}</ref> While AGGF1 is predominantly functional in [[Endothelial cells|endothelial]], [[vascular smooth muscle cells]], and [[Osteoblast|osteoblasts]], it also has activity in [[mast cells]], cardiac cells, [[Kupffer cell|Kupffer cells]] and [[hematopoietic stem cells]].<ref name="Fan_2009" /><ref name="Zhan_2016" /><ref name="Zhou_2016">{{cite journal | vauthors = Zhou B, Zeng S, Li L, Fan Z, Tian W, Li M, Xu H, Wu X, Fang M, Xu Y | title = Angiogenic factor with G patch and FHA domains 1 (Aggf1) regulates liver fibrosis by modulating TGF-β signaling | journal = Biochimica et Biophysica Acta | volume = 1862 | issue = 6 | pages = 1203–13 | date = June 2016 | pmid = 26850475 | doi = 10.1016/j.bbadis.2016.02.002 }}</ref><ref name="Tian_2004">{{cite journal | vauthors = Tian XL, Kadaba R, You SA, Liu M, Timur AA, Yang L, Chen Q, Szafranski P, Rao S, Wu L, Housman DE, DiCorleto PE, Driscoll DJ, Borrow J, Wang Q | title = Identification of an angiogenic factor that when mutated causes susceptibility to Klippel-Trenaunay syndrome | journal = Nature | volume = 427 | issue = 6975 | pages = 640–5 | date = February 2004 | pmid = 14961121 | pmc = 1618873 | doi = 10.1038/nature02320 }}</ref> AGGF1 mRNA has been detected in the heart, kidneys and limbs which indicates that the protein likely also functions in these organs.<ref name="Liu_2014" /> The proliferation of vascular smooth muscle cells is inhibited when AGGF1 is expressed.<ref name="Yao_2017b">{{cite journal | vauthors = Yao Y, Hu Z, Ye J, Hu C, Song Q, Da X, Yu Y, Li H, Xu C, Chen Q, Wang QK | title = Targeting AGGF1 (angiogenic factor with G patch and FHA domains 1) for Blocking Neointimal Formation After Vascular Injury | journal = Journal of the American Heart Association | volume = 6 | issue = 6 | date = June 2017 | pmid = 28649088 | pmc = 5669188 | doi = 10.1161/JAHA.117.005889 }}</ref> It has been found that AGGF1 is highly expressed in some malignant tumours which has implicated AGGF1 in cancer.<ref name="Yao_2017b" /> In vitro models have shown that AGGF1 localizes to cell periphery and directly outside of the cell.<ref name="Tian_2004" />  
AGGF1 is largely expressed during early embryonic vein specification, and expression is increased when endothelial cells are activated.<ref name="Liu_2014" /><ref name="Zhan_2016">{{cite journal | vauthors = Zhan M, Hori Y, Wada N, Ikeda J, Hata Y, Osuga K, Morii E | title = Angiogenic Factor with G-patch and FHA Domain 1 (AGGF1) Expression in Human Vascular Lesions | journal = Acta Histochemica et Cytochemica | volume = 49 | issue = 2 | pages = 75–81 | date = April 2016 | pmid = 27222614 | pmc = 4858542 | doi = 10.1267/ahc.15035 }}</ref> While AGGF1 is predominantly functional in [[Endothelial cells|endothelial]], [[vascular smooth muscle cells]], and [[osteoblast]]s, it also has activity in [[mast cells]], cardiac cells, [[Kupffer cell]]s and [[hematopoietic stem cells]].<ref name="Fan_2009" /><ref name="Zhan_2016" /><ref name="Zhou_2016">{{cite journal | vauthors = Zhou B, Zeng S, Li L, Fan Z, Tian W, Li M, Xu H, Wu X, Fang M, Xu Y | title = Angiogenic factor with G patch and FHA domains 1 (Aggf1) regulates liver fibrosis by modulating TGF-β signaling | journal = Biochimica et Biophysica Acta | volume = 1862 | issue = 6 | pages = 1203–13 | date = June 2016 | pmid = 26850475 | doi = 10.1016/j.bbadis.2016.02.002 }}</ref><ref name="Tian_2004">{{cite journal | vauthors = Tian XL, Kadaba R, You SA, Liu M, Timur AA, Yang L, Chen Q, Szafranski P, Rao S, Wu L, Housman DE, DiCorleto PE, Driscoll DJ, Borrow J, Wang Q | title = Identification of an angiogenic factor that when mutated causes susceptibility to Klippel-Trenaunay syndrome | journal = Nature | volume = 427 | issue = 6975 | pages = 640–5 | date = February 2004 | pmid = 14961121 | pmc = 1618873 | doi = 10.1038/nature02320 }}</ref> AGGF1 mRNA has been detected in the heart, kidneys and limbs which indicates that the protein likely also functions in these organs.<ref name="Liu_2014" /> The proliferation of vascular smooth muscle cells is inhibited when AGGF1 is expressed.<ref name="Yao_2017b">{{cite journal | vauthors = Yao Y, Hu Z, Ye J, Hu C, Song Q, Da X, Yu Y, Li H, Xu C, Chen Q, Wang QK | title = Targeting AGGF1 (angiogenic factor with G patch and FHA domains 1) for Blocking Neointimal Formation After Vascular Injury | journal = Journal of the American Heart Association | volume = 6 | issue = 6 | date = June 2017 | pmid = 28649088 | pmc = 5669188 | doi = 10.1161/JAHA.117.005889 | page=e005889}}</ref> It has been found that AGGF1 is highly expressed in some malignant tumours which has implicated AGGF1 in cancer.<ref name="Yao_2017b" /> In vitro models have shown that AGGF1 localizes to cell periphery and directly outside of the cell.<ref name="Tian_2004" />


Depending on the mutation type, AGGF1 mutations can be lethal in either the heterozygous or homozygous genotype due to its haploinsufficiency.<ref name="Liu_2014" /> Mice models have shown that heterozygous mutations can cause fatality due to [[Bleeding|hemorrhaging]] while homozygous mutations can prevent proper stem cell differentiation.<ref name="Liu_2014" />
Depending on the mutation type, AGGF1 mutations can be lethal in either the heterozygous or homozygous genotype due to its haploinsufficiency.<ref name="Liu_2014" /> Mice models have shown that heterozygous mutations can cause fatality due to [[Bleeding|hemorrhaging]] while homozygous mutations can prevent proper stem cell differentiation.<ref name="Liu_2014" />


== Homology ==
== Homology ==
Aggf1 is not unique to humans. This gene is [[Conserved sequence|conserved]] across many species, such as chimpanzees, Rhesus monkeys, dogs, cows, mice, rats, chickens, and frogs.<ref name="entrez" /> There are 212 organisms that have genes which are [[Homology (biology)|orthologs]] to AGGF1.<ref name="entrez" />
Aggf1 is not unique to humans. This gene is [[Conserved sequence|conserved]] across many species, such as chimpanzees, Rhesus monkeys, dogs, cows, mice, rats, chickens, and frogs.<ref name="entrez" /> There are 212 organisms that have genes which are [[Homology (biology)|orthologs]] to AGGF1.<ref name="entrez" />


Within the human chromosome, there are [[Pseudogene|pseudogenes]] related to AGGF1 are located on chromosomes 3, 4, 10 and 16 that have likely arisen due to [[Chromosomal translocation|translocation]] events.<ref name="entrez" />
Within the human chromosome, there are [[pseudogene]]s related to AGGF1 are located on chromosomes 3, 4, 10 and 16 that have likely arisen due to [[Chromosomal translocation|translocation]] events.<ref name="entrez" />


== Function ==
== Function ==
AGGF1 functions to regulate [[angiogenesis]] and vascular development.<ref name="Zhang_2016" />  [[Gene ontology]] has also implicated AGGF1 in cell adhesion, positive regulation of angiogenesis and endothelial cell proliferation.<ref name="entrez" /> Additionally, AGGF1 has been shown to protect against inflammation and ischemic injuries.<ref name="Zhou_2016" /> During embryongenesis, AGGF1 is required for hematopoietic stem cell specification and the differentiation of hematopoietic and endothelial cell lineages.<ref name="Liu_2014" /> Specifically, it regulates vascular endothelial [[cadherin]] (VE-cadherin) by inhibiting the phosphorylation of the cadherin and increasing its presence in the [[plasma membrane]] of endothelial cells.<ref name="Zhang_2016" /> AGGF1 is critical to the specification of veins and [[Hemangioblast|multipotent hemanigioblasts]], anti-inflammation, tumour angiogenesis, and inhibition of vascular permeability.<ref name="Lu_2016">{{cite journal | vauthors = Lu Q, Yao Y, Hu Z, Hu C, Song Q, Ye J, Xu C, Wang AZ, Chen Q, Wang QK | title = Angiogenic Factor AGGF1 Activates Autophagy with an Essential Role in Therapeutic Angiogenesis for Heart Disease | journal = PLoS Biology | volume = 14 | issue = 8 | pages = e1002529 | date = August 2016 | pmid = 27513923 | pmc = 4981375 | doi = 10.1371/journal.pbio.1002529 }}</ref> Additionally, it activates [[autophagy]] in specific cell types, such as [[endothelial]] cells, cardiac HL1 and H9C2 cells, and [[vascular smooth muscle cells]].<ref name="Zhang_2016" /><ref name="Liu_2014" /><ref name="Lu_2016" />
AGGF1 functions to regulate [[angiogenesis]] and vascular development.<ref name="Zhang_2016" /> [[Gene ontology]] has also implicated AGGF1 in cell adhesion, positive regulation of angiogenesis and endothelial cell proliferation.<ref name="entrez" /> Additionally, AGGF1 has been shown to protect against inflammation and ischemic injuries.<ref name="Zhou_2016" /> During embryongenesis, AGGF1 is required for hematopoietic stem cell specification and the differentiation of hematopoietic and endothelial cell lineages.<ref name="Liu_2014" /> Specifically, it regulates vascular endothelial [[cadherin]] (VE-cadherin) by inhibiting the phosphorylation of the cadherin and increasing its presence in the [[plasma membrane]] of endothelial cells.<ref name="Zhang_2016" /> AGGF1 is critical to the specification of veins and [[Hemangioblast|multipotent hemanigioblasts]], anti-inflammation, tumour angiogenesis, and inhibition of vascular permeability.<ref name="Lu_2016">{{cite journal | vauthors = Lu Q, Yao Y, Hu Z, Hu C, Song Q, Ye J, Xu C, Wang AZ, Chen Q, Wang QK | title = Angiogenic Factor AGGF1 Activates Autophagy with an Essential Role in Therapeutic Angiogenesis for Heart Disease | journal = PLoS Biology | volume = 14 | issue = 8 | pages = e1002529 | date = August 2016 | pmid = 27513923 | pmc = 4981375 | doi = 10.1371/journal.pbio.1002529 }}</ref> Additionally, it activates [[autophagy]] in specific cell types, such as [[endothelial]] cells, cardiac HL1 and H9C2 cells, and [[vascular smooth muscle cells]].<ref name="Zhang_2016" /><ref name="Liu_2014" /><ref name="Lu_2016" />


== Interactions ==
== Interactions ==
AGGF1 directly and indirectly interacts with many proteins. There are direct interactions between AGGF1 and [[TNFSF12]], another secreted angiogenic factor, that leads to increased angiogenesis.<ref name="Tian_2004" /> AGGF1 acts upstream of hemangioblast genes such as scl, fil1, and etsrp.<ref name="Li_2014" /> AGGF1 acts similarly to [[Vascular endothelial growth factor|VEGF]] - another gene implicated in vascular growth.<ref name="Li_2014" /> Additionally, AGGF1 is known to activate catalytic and regulatory subunits of [[Phosphoinositide 3-kinase|PI3K]].<ref name="Zhang_2016" /> This leads to downstream activation of [[AKT1|AKT]], [[GSK3B|GSK3b]] and [[P70-S6 Kinase 1|p70S6K]] signalling pathway which leads to vein specification and angiogenesis.<ref name="Zhang_2016" /><ref name="Li_2014" /> AGGF1 also interacts with vein specific markers such at [[FLT4|flt4]], [[DAB2|dab2]], and [[EPH receptor B4|ephB4]].<ref name="Chen_2013">{{cite journal | vauthors = Chen D, Li L, Tu X, Yin Z, Wang Q | title = Functional characterization of Klippel-Trenaunay syndrome gene AGGF1 identifies a novel angiogenic signaling pathway for specification of vein differentiation and angiogenesis during embryogenesis | journal = Human Molecular Genetics | volume = 22 | issue = 5 | pages = 963–76 | date = March 2013 | pmid = 23197652 | doi = 10.1093/hmg/dds501 }}</ref> [[CCL2|Ccl2]] has also been shown to interact with AGGF1 in [[Hepatocyte|hepatocytes]] through blocking [[NF-κB]]/p65 from binding to Ccl2.<ref>{{cite journal | vauthors = Xu W, Zeng S, Li M, Fan Z, Zhou B | title = Aggf1 attenuates hepatic inflammation and activation of hepatic stellate cells by repressing Ccl2 transcription | journal = Journal of Biomedical Research | volume = 31 | issue = 5 | pages = 428–436 | date = September 2017 | pmid = 28958996 | doi = 10.7555/JBR.30.20160046 }}</ref> AGGF1 activity is eliminated when Elk is overexpressed.<ref name="Yao_2017b" /> AGGF1 regulates autophagy by regulating expression of JNK genes.<ref name="Yao_2017b" />  [[SMAD (protein)|SMAD7]] and Aggf1 directly interact in the liver to inhibit fibrogenesis.<ref name="Zhou_2016" /> The presence of [[DNMT3B|DNMT3b]] will repress AGGF1 by acting on the promoter region of the gene.<ref name="Zhou_2016" />
AGGF1 directly and indirectly interacts with many proteins. There are direct interactions between AGGF1 and [[TNFSF12]], another secreted angiogenic factor, that leads to increased angiogenesis.<ref name="Tian_2004" /> AGGF1 acts upstream of hemangioblast genes such as scl, fil1, and etsrp.<ref name="Li_2014" /> AGGF1 acts similarly to [[Vascular endothelial growth factor|VEGF]] - another gene implicated in vascular growth.<ref name="Li_2014" /> Additionally, AGGF1 is known to activate catalytic and regulatory subunits of [[Phosphoinositide 3-kinase|PI3K]].<ref name="Zhang_2016" /> This leads to downstream activation of [[AKT1|AKT]], [[GSK3B|GSK3b]] and [[P70-S6 Kinase 1|p70S6K]] signalling pathway which leads to vein specification and angiogenesis.<ref name="Zhang_2016" /><ref name="Li_2014" /> AGGF1 also interacts with vein specific markers such at [[FLT4|flt4]], [[DAB2|dab2]], and [[EPH receptor B4|ephB4]].<ref name="Chen_2013">{{cite journal | vauthors = Chen D, Li L, Tu X, Yin Z, Wang Q | title = Functional characterization of Klippel-Trenaunay syndrome gene AGGF1 identifies a novel angiogenic signaling pathway for specification of vein differentiation and angiogenesis during embryogenesis | journal = Human Molecular Genetics | volume = 22 | issue = 5 | pages = 963–76 | date = March 2013 | pmid = 23197652 | doi = 10.1093/hmg/dds501 }}</ref> [[CCL2|Ccl2]] has also been shown to interact with AGGF1 in [[hepatocyte]]s through blocking [[NF-κB]]/p65 from binding to Ccl2.<ref>{{cite journal | vauthors = Xu W, Zeng S, Li M, Fan Z, Zhou B | title = Aggf1 attenuates hepatic inflammation and activation of hepatic stellate cells by repressing Ccl2 transcription | journal = Journal of Biomedical Research | volume = 31 | issue = 5 | pages = 428–436 | date = September 2017 | pmid = 28958996 | doi = 10.7555/JBR.30.20160046 | pmc=5706435}}</ref> AGGF1 activity is eliminated when Elk is overexpressed.<ref name="Yao_2017b" /> AGGF1 regulates autophagy by regulating expression of JNK genes.<ref name="Yao_2017b" /> [[SMAD (protein)|SMAD7]] and Aggf1 directly interact in the liver to inhibit fibrogenesis.<ref name="Zhou_2016" /> The presence of [[DNMT3B|DNMT3b]] will repress AGGF1 by acting on the promoter region of the gene.<ref name="Zhou_2016" />


== Clinical significance ==
== Clinical significance ==


=== Klippel-Trenaunay Syndrome ===
=== Klippel-Trenaunay Syndrome ===
Heterogeneous mutations in this gene causing deregulation of expression can lead to the vascular malformations associated with [[Klippel–Trénaunay syndrome|Klippel-Trenaunay syndrome]] (KTS).<ref name="Zhang_2016" /><ref name="Fan_2009" /><ref name="Chen_2013" /> Due to the haploinsufficient nature of AGGF1, individuals who have even one mutant allele may have KTS.<ref name="Zhang_2016" /> Studies done in mouse models have shown frequent [[haemorrhages]] and increased vascular permeability has been seen in mice who are [[heterozygous]] for Aggf1.<ref name="Zhang_2016" /> A translocation between the chromosome 5 q-arm at region 13 in band 3 and the chromosome 11 p-arm at region 15 in band 1 has been implicated in KTS.<ref name="Hu_2008" /> This translocation affects the AGGF1 promoter so there is a 3 fold increase in protein production.<ref name="Hu_2008" /> Single nucleotide polymorphisms in intron 11 and exon 7 were associated with KTS susceptibility even though neither of these SNPs resulted in an amino acid change.<ref name="Hu_2008" /> At one point, the E133K allele was thought to be a mutational hotspot - due to altered phosphorylation - causing KTS, but it has since been found as much as 3.3% of the population are carriers for the mutation.<ref name="Tian_2004" /><ref>{{cite journal | vauthors = Barker KT, Foulkes WD, Schwartz CE, Labadie C, Monsell F, Houlston RS, Harper J | title = Is the E133K allele of VG5Q associated with Klippel-Trenaunay and other overgrowth syndromes? | journal = Journal of Medical Genetics | volume = 43 | issue = 7 | pages = 613–4 | date = July 2006 | pmid = 16443853 | pmc = 2564558 | doi = 10.1136/jmg.2006.040790 }}</ref>
Heterogeneous mutations in this gene causing deregulation of expression can lead to the vascular malformations associated with [[Klippel–Trénaunay syndrome|Klippel-Trenaunay syndrome]] (KTS).<ref name="Zhang_2016" /><ref name="Fan_2009" /><ref name="Chen_2013" /> Due to the haploinsufficient nature of AGGF1, individuals who have even one mutant allele may have KTS.<ref name="Zhang_2016" /> Studies done in mouse models have shown frequent [[haemorrhages]] and increased vascular permeability has been seen in mice who are [[heterozygous]] for Aggf1.<ref name="Zhang_2016" /> A translocation between the chromosome 5 q-arm at region 13 in band 3 and the chromosome 11 p-arm at region 15 in band 1 has been implicated in KTS.<ref name="pmid18564129"/> This translocation affects the AGGF1 promoter so there is a 3 fold increase in protein production.<ref name="pmid18564129"/> Single nucleotide polymorphisms in intron 11 and exon 7 were associated with KTS susceptibility even though neither of these SNPs resulted in an amino acid change.<ref name="pmid18564129"/> At one point, the E133K allele was thought to be a mutational hotspot - due to altered phosphorylation - causing KTS, but it has since been found as much as 3.3% of the population are carriers for the mutation.<ref name="Tian_2004" /><ref>{{cite journal | vauthors = Barker KT, Foulkes WD, Schwartz CE, Labadie C, Monsell F, Houlston RS, Harper J | title = Is the E133K allele of VG5Q associated with Klippel-Trenaunay and other overgrowth syndromes? | journal = Journal of Medical Genetics | volume = 43 | issue = 7 | pages = 613–4 | date = July 2006 | pmid = 16443853 | pmc = 2564558 | doi = 10.1136/jmg.2006.040790 }}</ref>


=== Heart Disease ===
=== Heart Disease ===
AGGF1 has also been implicated in treatment after vascular smooth muscle cell damage due to [[coronary artery disease]] and [[myocardial infarction]].<ref name="Yao_2017b" /> By blocking vascular permeability and regulating vascular smooth muscle cell phenotypic switching, AGGF1 protein therapy is currently being investigated as a new method of treating both of these diseases.<ref name="Yao_2017b" />
AGGF1 has also been implicated in treatment after vascular smooth muscle cell damage due to [[coronary artery disease]] and [[myocardial infarction]].<ref name="Yao_2017b" /> By blocking vascular permeability and regulating vascular smooth muscle cell phenotypic switching, AGGF1 protein therapy is currently being investigated as a new method of treating both of these diseases.<ref name="Yao_2017b" />


=== Cancer ===
=== Cancer ===
Abberant AGGF1 has been implicated in multiple cancers and functions in tumour initiation and progression.<ref name="Wang_2015" /> For example, both hepatocellular carcinoma and gastric cancer survivability is related to the levels of AGGF1 expression in tumours.<ref name="Yao_2017" /><ref name="Wang_2015" /> AGGF1 has been found to have higher expression in tumours than the surrounding tissues, and higher levels of AGGF1 are associated with a poor patient prognosis.<ref name="Yao_2017" /><ref name="Wang_2015" />
Aberrant AGGF1 has been implicated in multiple cancers and functions in tumour initiation and progression.<ref name="Wang_2015" /> For example, both hepatocellular carcinoma and gastric cancer survivability is related to the levels of AGGF1 expression in tumours.<ref name="Yao_2017" /><ref name="Wang_2015" /> AGGF1 has been found to have higher expression in tumours than the surrounding tissues, and higher levels of AGGF1 are associated with a poor patient prognosis.<ref name="Yao_2017" /><ref name="Wang_2015" />


== See also ==
== See also ==
Line 63: Line 64:
== External links ==
== External links ==
* {{UCSC gene info|AGGF1}}
* {{UCSC gene info|AGGF1}}
{{gene-5-stub}}

Latest revision as of 17:27, 10 October 2018

VALUE_ERROR (nil)
Identifiers
Aliases
External IDsGeneCards: [1]
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

n/a

RefSeq (protein)

n/a

n/a

Location (UCSC)n/an/a
PubMed searchn/an/a
Wikidata
View/Edit Human

Angiogenic factor with G patch and FHA domains 1 is a protein that in humans is encoded by the AGGF1 gene.[1][2][3]

AGGF1 is a human gene that functions as an angiogenic factor with a G-patch and forkhead-associated domain.[4] This gene is predominantly expressed in activated, plump endothelial cells and acts to regulate angiogenesis and vascular development.[5] AGGF1 is known to interact with a wide range of proteins involved in vascular development.[6] Mutations to AGGF1 have been implicated in multiple cancers and is known to cause the rare congenital condition, Klippel-Trenaunay syndrome.[5][7][8]

Gene

The gene was originally named VG5Q, indicating that it was a vascular gene on chromosome 5, but the name was later changed to reflect its function, instead of just its location.[9]

The AGGF1 gene promoter does not contain a TATA box and contains 2 transcription start sites that are -367 and -364 base pairs ahead of the base translation start site.[9] The gene promoter contains over 50 CpG islands, which makes it a DNA methylation target.[9] AGGF1 is regulated by 2 repressor sites and 2 activator sites.[9] While the presence of 2 repressor and 2 activator sites is clear, the only known transcription factor that regulates AGGF1 is GATA1.[9] GATA1 binds upstream of the AGGF1 gene promoter at -295 and -300, and the binding of GATA1 will lead to increased AGGF1 expression.[5][9] For the gene to be fully expressed, both of the activator sites must be bound by the transcription factors, GATA1 and another unknown factor.[9]

Protein

To form a protein, an mRNA transcript must be transcribed from the DNA. For AGGF1, the mRNA transcript contains 14 exons and 34 807 nucleotides.[1]

There are 714 amino acids present in this protein, and it has a molecular weight of 80997 Da.[10] It contains a coiled coil domain at positions 18-88 and an OCRE domain at the N terminus.[10] The G-patch domain is located at amino acids 619-663 while the forkhead-associated domain is located at amino acids 435-508.[10] While it is known that these domains are present in the protein, their role in protein function remains unclear.

AGGF1 was the third haploinsufficient human gene identified.[5] Haploinsufficiency means AGGF1 is "dose dependent" so any reductions in protein product can have phenotypic consequences on the vascular development of the organism.

Expression

AGGF1 is largely expressed during early embryonic vein specification, and expression is increased when endothelial cells are activated.[10][4] While AGGF1 is predominantly functional in endothelial, vascular smooth muscle cells, and osteoblasts, it also has activity in mast cells, cardiac cells, Kupffer cells and hematopoietic stem cells.[9][4][11][12] AGGF1 mRNA has been detected in the heart, kidneys and limbs which indicates that the protein likely also functions in these organs.[10] The proliferation of vascular smooth muscle cells is inhibited when AGGF1 is expressed.[13] It has been found that AGGF1 is highly expressed in some malignant tumours which has implicated AGGF1 in cancer.[13] In vitro models have shown that AGGF1 localizes to cell periphery and directly outside of the cell.[12]

Depending on the mutation type, AGGF1 mutations can be lethal in either the heterozygous or homozygous genotype due to its haploinsufficiency.[10] Mice models have shown that heterozygous mutations can cause fatality due to hemorrhaging while homozygous mutations can prevent proper stem cell differentiation.[10]

Homology

Aggf1 is not unique to humans. This gene is conserved across many species, such as chimpanzees, Rhesus monkeys, dogs, cows, mice, rats, chickens, and frogs.[3] There are 212 organisms that have genes which are orthologs to AGGF1.[3]

Within the human chromosome, there are pseudogenes related to AGGF1 are located on chromosomes 3, 4, 10 and 16 that have likely arisen due to translocation events.[3]

Function

AGGF1 functions to regulate angiogenesis and vascular development.[5] Gene ontology has also implicated AGGF1 in cell adhesion, positive regulation of angiogenesis and endothelial cell proliferation.[3] Additionally, AGGF1 has been shown to protect against inflammation and ischemic injuries.[11] During embryongenesis, AGGF1 is required for hematopoietic stem cell specification and the differentiation of hematopoietic and endothelial cell lineages.[10] Specifically, it regulates vascular endothelial cadherin (VE-cadherin) by inhibiting the phosphorylation of the cadherin and increasing its presence in the plasma membrane of endothelial cells.[5] AGGF1 is critical to the specification of veins and multipotent hemanigioblasts, anti-inflammation, tumour angiogenesis, and inhibition of vascular permeability.[14] Additionally, it activates autophagy in specific cell types, such as endothelial cells, cardiac HL1 and H9C2 cells, and vascular smooth muscle cells.[5][10][14]

Interactions

AGGF1 directly and indirectly interacts with many proteins. There are direct interactions between AGGF1 and TNFSF12, another secreted angiogenic factor, that leads to increased angiogenesis.[12] AGGF1 acts upstream of hemangioblast genes such as scl, fil1, and etsrp.[6] AGGF1 acts similarly to VEGF - another gene implicated in vascular growth.[6] Additionally, AGGF1 is known to activate catalytic and regulatory subunits of PI3K.[5] This leads to downstream activation of AKT, GSK3b and p70S6K signalling pathway which leads to vein specification and angiogenesis.[5][6] AGGF1 also interacts with vein specific markers such at flt4, dab2, and ephB4.[15] Ccl2 has also been shown to interact with AGGF1 in hepatocytes through blocking NF-κB/p65 from binding to Ccl2.[16] AGGF1 activity is eliminated when Elk is overexpressed.[13] AGGF1 regulates autophagy by regulating expression of JNK genes.[13] SMAD7 and Aggf1 directly interact in the liver to inhibit fibrogenesis.[11] The presence of DNMT3b will repress AGGF1 by acting on the promoter region of the gene.[11]

Clinical significance

Klippel-Trenaunay Syndrome

Heterogeneous mutations in this gene causing deregulation of expression can lead to the vascular malformations associated with Klippel-Trenaunay syndrome (KTS).[5][9][15] Due to the haploinsufficient nature of AGGF1, individuals who have even one mutant allele may have KTS.[5] Studies done in mouse models have shown frequent haemorrhages and increased vascular permeability has been seen in mice who are heterozygous for Aggf1.[5] A translocation between the chromosome 5 q-arm at region 13 in band 3 and the chromosome 11 p-arm at region 15 in band 1 has been implicated in KTS.[1] This translocation affects the AGGF1 promoter so there is a 3 fold increase in protein production.[1] Single nucleotide polymorphisms in intron 11 and exon 7 were associated with KTS susceptibility even though neither of these SNPs resulted in an amino acid change.[1] At one point, the E133K allele was thought to be a mutational hotspot - due to altered phosphorylation - causing KTS, but it has since been found as much as 3.3% of the population are carriers for the mutation.[12][17]

Heart Disease

AGGF1 has also been implicated in treatment after vascular smooth muscle cell damage due to coronary artery disease and myocardial infarction.[13] By blocking vascular permeability and regulating vascular smooth muscle cell phenotypic switching, AGGF1 protein therapy is currently being investigated as a new method of treating both of these diseases.[13]

Cancer

Aberrant AGGF1 has been implicated in multiple cancers and functions in tumour initiation and progression.[8] For example, both hepatocellular carcinoma and gastric cancer survivability is related to the levels of AGGF1 expression in tumours.[7][8] AGGF1 has been found to have higher expression in tumours than the surrounding tissues, and higher levels of AGGF1 are associated with a poor patient prognosis.[7][8]

See also

References

  1. 1.0 1.1 1.2 1.3 1.4 Hu Y, Li L, Seidelmann SB, Timur AA, Shen PH, Driscoll DJ, Wang QK (September 2008). "Identification of association of common AGGF1 variants with susceptibility for Klippel-Trenaunay syndrome using the structure association program". Annals of Human Genetics. 72 (Pt 5): 636–43. doi:10.1111/j.1469-1809.2008.00458.x. PMC 2602961. PMID 18564129.
  2. Gutierrez S, Magano L, Delicado A, Mori MA, de Torres ML, Fernández L, Palomares M, Fernández E, Tarduchy GR, Molano J, Gracia R, Pajares IL, Lapunzina P (December 2006). "The G397A (E133K) change in the AGGF1 (VG5Q) gene is a single nucleotide polymorphism in the Spanish population". American Journal of Medical Genetics. Part A. 140 (24): 2832–3. doi:10.1002/ajmg.a.31532. PMID 17103452.
  3. 3.0 3.1 3.2 3.3 3.4 "Entrez Gene: AGGF1 angiogenic factor with G patch and FHA domains 1".
  4. 4.0 4.1 4.2 Zhan M, Hori Y, Wada N, Ikeda J, Hata Y, Osuga K, Morii E (April 2016). "Angiogenic Factor with G-patch and FHA Domain 1 (AGGF1) Expression in Human Vascular Lesions". Acta Histochemica et Cytochemica. 49 (2): 75–81. doi:10.1267/ahc.15035. PMC 4858542. PMID 27222614.
  5. 5.00 5.01 5.02 5.03 5.04 5.05 5.06 5.07 5.08 5.09 5.10 5.11 Zhang T, Yao Y, Wang J, Li Y, He P, Pasupuleti V, Hu Z, Jia X, Song Q, Tian XL, Hu C, Chen Q, Wang QK (December 2016). "Haploinsufficiency of Klippel-Trenaunay syndrome gene Aggf1 inhibits developmental and pathological angiogenesis by inactivating PI3K and AKT and disrupts vascular integrity by activating VE-cadherin". Human Molecular Genetics. 25 (23): 5094–5110. doi:10.1093/hmg/ddw273. PMC 6078640. PMID 27522498.
  6. 6.0 6.1 6.2 6.3 Li L, Chen D, Li J, Wang X, Wang N, Xu C, Wang QK (January 2014). "Aggf1 acts at the top of the genetic regulatory hierarchy in specification of hemangioblasts in zebrafish". Blood. 123 (4): 501–8. doi:10.1182/blood-2013-07-514612. PMC 3901065. PMID 24277077.
  7. 7.0 7.1 7.2 Yao HH, Wang BJ, Wu Y, Huang Q (2017). "High Expression of Angiogenic Factor with G-Patch and FHA Domain1 (AGGF1) Predicts Poor Prognosis in Gastric Cancer". Medical Science Monitor : International Medical Journal of Experimental and Clinical Research. 23: 1286–1294. doi:10.12659/msm.903248. PMC 5362190. PMID 28289272.
  8. 8.0 8.1 8.2 8.3 Wang W, Li GY, Zhu JY, Huang DB, Zhou HC, Zhong W, Ji CS (April 2015). "Overexpression of AGGF1 is correlated with angiogenesis and poor prognosis of hepatocellular carcinoma". Medical Oncology. 32 (4): 131. doi:10.1007/s12032-015-0574-2. PMID 25796501.
  9. 9.0 9.1 9.2 9.3 9.4 9.5 9.6 9.7 9.8 Fan C, Ouyang P, Timur AA, He P, You SA, Hu Y, Ke T, Driscoll DJ, Chen Q, Wang QK (August 2009). "Novel roles of GATA1 in regulation of angiogenic factor AGGF1 and endothelial cell function". The Journal of Biological Chemistry. 284 (35): 23331–43. doi:10.1074/jbc.M109.036079. PMC 2749107. PMID 19556247.
  10. 10.0 10.1 10.2 10.3 10.4 10.5 10.6 10.7 10.8 Liu Y, Yang H, Song L, Li N, Han QY, Tian C, Gao E, Du J, Xia YL, Li HH (August 2014). "AGGF1 protects from myocardial ischemia/reperfusion injury by regulating myocardial apoptosis and angiogenesis". Apoptosis. 19 (8): 1254–68. doi:10.1007/s10495-014-1001-4. PMID 24893993.
  11. 11.0 11.1 11.2 11.3 Zhou B, Zeng S, Li L, Fan Z, Tian W, Li M, Xu H, Wu X, Fang M, Xu Y (June 2016). "Angiogenic factor with G patch and FHA domains 1 (Aggf1) regulates liver fibrosis by modulating TGF-β signaling". Biochimica et Biophysica Acta. 1862 (6): 1203–13. doi:10.1016/j.bbadis.2016.02.002. PMID 26850475.
  12. 12.0 12.1 12.2 12.3 Tian XL, Kadaba R, You SA, Liu M, Timur AA, Yang L, Chen Q, Szafranski P, Rao S, Wu L, Housman DE, DiCorleto PE, Driscoll DJ, Borrow J, Wang Q (February 2004). "Identification of an angiogenic factor that when mutated causes susceptibility to Klippel-Trenaunay syndrome". Nature. 427 (6975): 640–5. doi:10.1038/nature02320. PMC 1618873. PMID 14961121.
  13. 13.0 13.1 13.2 13.3 13.4 13.5 Yao Y, Hu Z, Ye J, Hu C, Song Q, Da X, Yu Y, Li H, Xu C, Chen Q, Wang QK (June 2017). "Targeting AGGF1 (angiogenic factor with G patch and FHA domains 1) for Blocking Neointimal Formation After Vascular Injury". Journal of the American Heart Association. 6 (6): e005889. doi:10.1161/JAHA.117.005889. PMC 5669188. PMID 28649088.
  14. 14.0 14.1 Lu Q, Yao Y, Hu Z, Hu C, Song Q, Ye J, Xu C, Wang AZ, Chen Q, Wang QK (August 2016). "Angiogenic Factor AGGF1 Activates Autophagy with an Essential Role in Therapeutic Angiogenesis for Heart Disease". PLoS Biology. 14 (8): e1002529. doi:10.1371/journal.pbio.1002529. PMC 4981375. PMID 27513923.
  15. 15.0 15.1 Chen D, Li L, Tu X, Yin Z, Wang Q (March 2013). "Functional characterization of Klippel-Trenaunay syndrome gene AGGF1 identifies a novel angiogenic signaling pathway for specification of vein differentiation and angiogenesis during embryogenesis". Human Molecular Genetics. 22 (5): 963–76. doi:10.1093/hmg/dds501. PMID 23197652.
  16. Xu W, Zeng S, Li M, Fan Z, Zhou B (September 2017). "Aggf1 attenuates hepatic inflammation and activation of hepatic stellate cells by repressing Ccl2 transcription". Journal of Biomedical Research. 31 (5): 428–436. doi:10.7555/JBR.30.20160046. PMC 5706435. PMID 28958996.
  17. Barker KT, Foulkes WD, Schwartz CE, Labadie C, Monsell F, Houlston RS, Harper J (July 2006). "Is the E133K allele of VG5Q associated with Klippel-Trenaunay and other overgrowth syndromes?". Journal of Medical Genetics. 43 (7): 613–4. doi:10.1136/jmg.2006.040790. PMC 2564558. PMID 16443853.

Further reading

External links