MAP4K4: Difference between revisions

Jump to navigation Jump to search
m (task, replaced: Current Biology : CB → Current Biology, removed stub tag using AWB)
imported>KolbertBot
m (Bot: HTTP→HTTPS (v485))
 
Line 2: Line 2:
'''Mitogen-activated protein kinase kinase kinase kinase 4''' (MAP4K4) – also known as hepatocyte progenitor kinase-like/germinal center kinase-like kinase (HGK) and Nck-interacting kinase (NIK) – is an enzyme, specifically a serine/threonine (S/T) [[kinase]] encoded by the MAP4K4 gene in humans.<ref name="Delpire_2009" /><ref name="Gao_2016" />
'''Mitogen-activated protein kinase kinase kinase kinase 4''' (MAP4K4) – also known as hepatocyte progenitor kinase-like/germinal center kinase-like kinase (HGK) and Nck-interacting kinase (NIK) – is an enzyme, specifically a serine/threonine (S/T) [[kinase]] encoded by the MAP4K4 gene in humans.<ref name="Delpire_2009" /><ref name="Gao_2016" />


MAP4K4 is involved in a wide array of physiological processes including cell migration, proliferation and adhesion;<ref name="Buburuzan_2011" /> its activity has been implicated in systemic [[inflammation]],<ref>{{cite journal | vauthors = Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP | title = Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation | journal = Nature | volume = 458 | issue = 7242 | pages = 1180–4 | date = April 2009 | pmid = 19407801 | doi = 10.1038/nature07774 }}</ref> [[metabolic disorder]]s,<ref name="Virbasius_2016">{{cite journal | vauthors = Virbasius JV, Czech MP | title = Map4k4 Signaling Nodes in Metabolic and Cardiovascular Diseases | journal = Trends in Endocrinology and Metabolism | volume = 27 | issue = 7 | pages = 484–92 | date = July 2016 | pmid = 27160798 | doi = 10.1016/j.tem.2016.04.006 }}</ref> [[cardiovascular disease]]<ref name="Virbasius_2016" /> and [[cancer]].<ref name="Gao_2016" />
MAP4K4 is involved in a wide array of physiological processes including cell migration, proliferation and adhesion;<ref name="Buburuzan_2011" /> its activity has been implicated in systemic [[inflammation]],<ref>{{cite journal | vauthors = Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP | title = Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation | journal = Nature | volume = 458 | issue = 7242 | pages = 1180–4 | date = April 2009 | pmid = 19407801 | doi = 10.1038/nature07774 | pmc=2879154}}</ref> [[metabolic disorder]]s,<ref name="Virbasius_2016">{{cite journal | vauthors = Virbasius JV, Czech MP | title = Map4k4 Signaling Nodes in Metabolic and Cardiovascular Diseases | journal = Trends in Endocrinology and Metabolism | volume = 27 | issue = 7 | pages = 484–92 | date = July 2016 | pmid = 27160798 | doi = 10.1016/j.tem.2016.04.006 | pmc=4912878}}</ref> [[cardiovascular disease]]<ref name="Virbasius_2016" /> and [[cancer]].<ref name="Gao_2016" />


While MAP4K4 has been found to be upregulated in a wide array of cancers,  there is currently limited information regarding its specific involvement. However, there is increasing evidence that suggests MAP4K4 has an important role in the development and progression of cancer, and may serve as a novel target for cancer therapeutics.<ref name="Gao_2016" />
While MAP4K4 has been found to be upregulated in a wide array of cancers,  there is currently limited information regarding its specific involvement. However, there is increasing evidence that suggests MAP4K4 has an important role in the development and progression of cancer, and may serve as a novel target for cancer therapeutics.<ref name="Gao_2016" />


== Discovery and classification ==
== Discovery and classification ==
MAP4K4 is categorized under the mammalian sterile 20 protein (Ste20p) kinase family due to its shared homology with the Ste20p kinase found in budding yeast<ref name="Delpire_2009">{{cite journal | vauthors = Delpire E | title = The mammalian family of sterile 20p-like protein kinases | journal = Pflugers Archiv | volume = 458 | issue = 5 | pages = 953–67 | date = September 2009 | pmid = 19399514 | doi = 10.1007/s00424-009-0674-y }}</ref> and is a member of the GCK-IV subfamily. Mammalian MAP4K4 was initially identified in mice as a kinase activator for a protein called Nck<ref name="Su_1997">{{cite journal | vauthors = Su YC, Han J, Xu S, Cobb M, Skolnik EY | title = NIK is a new Ste20-related kinase that binds NCK and MEKK1 and activates the SAPK/JNK cascade via a conserved regulatory domain | journal = The EMBO Journal | volume = 16 | issue = 6 | pages = 1279–90 | date = March 1997 | pmid = 9135144 | doi = 10.1093/emboj/16.6.1279 }}</ref> followed shortly by identification and cloning of the human orthologue encoded by the MAP4K4 gene.<ref name="Yao_1999">{{cite journal | vauthors = Yao Z, Zhou G, Wang XS, Brown A, Diener K, Gan H, Tan TH | title = A novel human STE20-related protein kinase, HGK, that specifically activates the c-Jun N-terminal kinase signaling pathway | journal = The Journal of Biological Chemistry | volume = 274 | issue = 4 | pages = 2118–25 | date = January 1999 | pmid = 9890973 | doi = 10.1074/jbc.274.4.2118 }}</ref>
MAP4K4 is categorized under the mammalian sterile 20 protein (Ste20p) kinase family due to its shared homology with the Ste20p kinase found in budding yeast<ref name="Delpire_2009">{{cite journal | vauthors = Delpire E | title = The mammalian family of sterile 20p-like protein kinases | journal = Pflügers Archiv | volume = 458 | issue = 5 | pages = 953–67 | date = September 2009 | pmid = 19399514 | doi = 10.1007/s00424-009-0674-y }}</ref> and is a member of the GCK-IV subfamily. Mammalian MAP4K4 was initially identified in mice as a kinase activator for a protein called Nck<ref name="Su_1997">{{cite journal | vauthors = Su YC, Han J, Xu S, Cobb M, Skolnik EY | title = NIK is a new Ste20-related kinase that binds NCK and MEKK1 and activates the SAPK/JNK cascade via a conserved regulatory domain | journal = The EMBO Journal | volume = 16 | issue = 6 | pages = 1279–90 | date = March 1997 | pmid = 9135144 | doi = 10.1093/emboj/16.6.1279 | pmc=1169726}}</ref> followed shortly by identification and cloning of the human orthologue encoded by the MAP4K4 gene.<ref name="Yao_1999">{{cite journal | vauthors = Yao Z, Zhou G, Wang XS, Brown A, Diener K, Gan H, Tan TH | title = A novel human STE20-related protein kinase, HGK, that specifically activates the c-Jun N-terminal kinase signaling pathway | journal = The Journal of Biological Chemistry | volume = 274 | issue = 4 | pages = 2118–25 | date = January 1999 | pmid = 9890973 | doi = 10.1074/jbc.274.4.2118 }}</ref>


== Structure and expression ==
== Structure and expression ==
Line 17: Line 17:
* C-Terminal Hydrophobic Leucine-Rich Citron Homology Domain (CNH)
* C-Terminal Hydrophobic Leucine-Rich Citron Homology Domain (CNH)
* Interdomain - Connects the kinase and CNH domains, facilitates protein-protein interactions. Although it has been identified, its structural components and functionality are currently poorly understood
* Interdomain - Connects the kinase and CNH domains, facilitates protein-protein interactions. Although it has been identified, its structural components and functionality are currently poorly understood
[[File:MAP4K4_V2.jpg|link=https://en.wikipedia.org/wiki/File:MAP4K4_V2.jpg|thumb|550x550px|'''Figure 1.''' Schematic representation of MAP4K4 structure, depicts the N-terminal kinase domain, C-terminal citron homology domain (regulatory function) and the interdomain (facilitates protein interactions).<ref name="Gao_2016">{{cite journal | vauthors = Gao X, Gao C, Liu G, Hu J | title = MAP4K4: an emerging therapeutic target in cancer | journal = Cell & Bioscience | volume = 6 | pages = 56 | date = 2016-10-28 | pmid = 27800153 | doi = 10.1186/s13578-016-0121-7 }}</ref><ref>{{Cite web|url=http://www.uniprot.org/uniprot/A0A0D9SG62|title=MAP4K4 - Mitogen-activated protein kinase kinase kinase kinase 4 - Homo sapiens (Human) - MAP4K4 gene & protein|website=www.uniprot.org|access-date=2017-11-30}}</ref> Note: the ''numbers'' indicate amino acid positions.]]
[[File:MAP4K4_V2.jpg|link=https://en.wikipedia.org/wiki/File:MAP4K4_V2.jpg|thumb|550x550px|'''Figure 1.''' Schematic representation of MAP4K4 structure, depicts the N-terminal kinase domain, C-terminal citron homology domain (regulatory function) and the interdomain (facilitates protein interactions).<ref name="Gao_2016">{{cite journal | vauthors = Gao X, Gao C, Liu G, Hu J | title = MAP4K4: an emerging therapeutic target in cancer | journal = Cell & Bioscience | volume = 6 | pages = 56 | date = 2016-10-28 | pmid = 27800153 | doi = 10.1186/s13578-016-0121-7 | pmc=5084373}}</ref><ref>{{Cite web|url=https://www.uniprot.org/uniprot/A0A0D9SG62|title=MAP4K4 - Mitogen-activated protein kinase kinase kinase kinase 4 - Homo sapiens (Human) - MAP4K4 gene & protein|website=www.uniprot.org|access-date=2017-11-30}}</ref> Note: the ''numbers'' indicate amino acid positions.]]
[[Alternative splicing]] of the MAP4K4 gene yields five functional [[Protein isoform|isoforms]], all of which display fully homologous kinase and CNH domains but differences in the interdomain domain.<ref name="pmid25625039">{{cite journal | vauthors = Santhana Kumar K, Tripolitsioti D, Ma M, Grählert J, Egli KB, Fiaschetti G, Shalaby T, Grotzer MA, Baumgartner M | title = The Ser/Thr kinase MAP4K4 drives c-Met-induced motility and invasiveness in a cell-based model of SHH medulloblastoma | journal = SpringerPlus | volume = 4 | issue = | pages = 19 | year = 2015 | pmid = 25625039 | pmc = 4302160 | doi = 10.1186/s40064-015-0784-2 }}</ref> While the biological significance of these isoforms remains to be determined, it can be speculated that such variations alter and determine MAP4K4's interactions with other proteins and factors, ultimately leading to the activation/inhibition of different biochemical and physiological cascades.
[[Alternative splicing]] of the MAP4K4 gene yields five functional [[Protein isoform|isoforms]], all of which display fully homologous kinase and CNH domains but differences in the interdomain domain.<ref name="pmid25625039">{{cite journal | vauthors = Santhana Kumar K, Tripolitsioti D, Ma M, Grählert J, Egli KB, Fiaschetti G, Shalaby T, Grotzer MA, Baumgartner M | title = The Ser/Thr kinase MAP4K4 drives c-Met-induced motility and invasiveness in a cell-based model of SHH medulloblastoma | journal = SpringerPlus | volume = 4 | issue = | pages = 19 | year = 2015 | pmid = 25625039 | pmc = 4302160 | doi = 10.1186/s40064-015-0784-2 }}</ref> While the biological significance of these isoforms remains to be determined, it can be speculated that such variations alter and determine MAP4K4's interactions with other proteins and factors, ultimately leading to the activation/inhibition of different biochemical and physiological cascades.


The mammalian class of Ste20 kinases require phosphorylation at specific sites for full activity. Primary phosphorylation at the activation site in their kinase domain is believed to cause a conformational change in the protein, stabilizing the structure of its activation segment to allow suitable substrate binding.<ref name="Delpire_2009" /> Secondary sites also require phosphorylation for the enzyme to assume full activation and is achieved via autophosphorylation or by upstream kinases.<ref name="Delpire_2009" />
The mammalian class of Ste20 kinases require phosphorylation at specific sites for full activity. Primary phosphorylation at the activation site in their kinase domain is believed to cause a conformational change in the protein, stabilizing the structure of its activation segment to allow suitable substrate binding.<ref name="Delpire_2009" /> Secondary sites also require phosphorylation for the enzyme to assume full activation and is achieved via autophosphorylation or by upstream kinases.<ref name="Delpire_2009" />


To date, MAP4K4 has been found to be expressed in all tissue types<ref name="Yao_1999" /> with a relatively more pronounced expression in the brain and testes.<ref name="Wright_2003">{{cite journal | vauthors = Wright JH, Wang X, Manning G, LaMere BJ, Le P, Zhu S, Khatry D, Flanagan PM, Buckley SD, Whyte DB, Howlett AR, Bischoff JR, Lipson KE, Jallal B | title = The STE20 kinase HGK is broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion | journal = Molecular and Cellular Biology | volume = 23 | issue = 6 | pages = 2068–82 | date = March 2003 | pmid = 12612079 | doi = 10.1128/mcb.23.6.2068-2082.2003 }}</ref> Multiple isoforms of MAP4K4 can be present at any given time in the same cell but the abundance of each isoform in the cell differs depending on the cell-type or tissue-type.<ref name="Wright_2003" />
To date, MAP4K4 has been found to be expressed in all tissue types<ref name="Yao_1999" /> with a relatively more pronounced expression in the brain and testes.<ref name="Wright_2003">{{cite journal | vauthors = Wright JH, Wang X, Manning G, LaMere BJ, Le P, Zhu S, Khatry D, Flanagan PM, Buckley SD, Whyte DB, Howlett AR, Bischoff JR, Lipson KE, Jallal B | title = The STE20 kinase HGK is broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion | journal = Molecular and Cellular Biology | volume = 23 | issue = 6 | pages = 2068–82 | date = March 2003 | pmid = 12612079 | doi = 10.1128/mcb.23.6.2068-2082.2003 | pmc=149462}}</ref> Multiple isoforms of MAP4K4 can be present at any given time in the same cell but the abundance of each isoform in the cell differs depending on the cell-type or tissue-type.<ref name="Wright_2003" />
* E.g. In humans, the shorter isoform of MAP4K4 is predominantly expressed in organs including the liver, placenta, skeletal muscles while a longer isoform is expressed in the brain
* E.g. In humans, the shorter isoform of MAP4K4 is predominantly expressed in organs including the liver, placenta, skeletal muscles while a longer isoform is expressed in the brain


Line 42: Line 42:
MAP4K4 has been identified to be involved in the negative regulation of insulin-dependent glucose transport. There is increasing evidence suggesting cytokines such as TNF-α mediate biological effects antagonistic to insulin action and induce inflammation observed in obesity.<ref>{{cite journal | vauthors = Hotamisligil GS, Spiegelman BM | title = Tumor necrosis factor alpha: a key component of the obesity-diabetes link | journal = Diabetes | volume = 43 | issue = 11 | pages = 1271–8 | date = November 1994 | pmid = 7926300 | doi = 10.2337/diab.43.11.1271 }}</ref><ref name="pmid8899294">{{cite journal | vauthors = Skolnik EY, Marcusohn J | title = Inhibition of insulin receptor signaling by TNF: potential role in obesity and non-insulin-dependent diabetes mellitus | journal = Cytokine & Growth Factor Reviews | volume = 7 | issue = 2 | pages = 161–73 | year = 1996 | pmid = 8899294 | doi = 10.1016/1359-6101(96)00021-4 }}</ref> TNF-α specifically attenuates the signaling pathway initiated by insulin receptors, reducing the amount of glucose transport and uptake;<ref>{{cite journal | vauthors = Peraldi P, Hotamisligil GS, Buurman WA, White MF, Spiegelman BM | title = Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase | journal = The Journal of Biological Chemistry | volume = 271 | issue = 22 | pages = 13018–22 | date = May 1996 | pmid = 8662983 | doi = 10.1074/jbc.271.22.13018 }}</ref> and it is believed that MAP4K4 functions as an upstream signaling element in the TNF-α signaling cascade.<ref name="Yao_1999" />
MAP4K4 has been identified to be involved in the negative regulation of insulin-dependent glucose transport. There is increasing evidence suggesting cytokines such as TNF-α mediate biological effects antagonistic to insulin action and induce inflammation observed in obesity.<ref>{{cite journal | vauthors = Hotamisligil GS, Spiegelman BM | title = Tumor necrosis factor alpha: a key component of the obesity-diabetes link | journal = Diabetes | volume = 43 | issue = 11 | pages = 1271–8 | date = November 1994 | pmid = 7926300 | doi = 10.2337/diab.43.11.1271 }}</ref><ref name="pmid8899294">{{cite journal | vauthors = Skolnik EY, Marcusohn J | title = Inhibition of insulin receptor signaling by TNF: potential role in obesity and non-insulin-dependent diabetes mellitus | journal = Cytokine & Growth Factor Reviews | volume = 7 | issue = 2 | pages = 161–73 | year = 1996 | pmid = 8899294 | doi = 10.1016/1359-6101(96)00021-4 }}</ref> TNF-α specifically attenuates the signaling pathway initiated by insulin receptors, reducing the amount of glucose transport and uptake;<ref>{{cite journal | vauthors = Peraldi P, Hotamisligil GS, Buurman WA, White MF, Spiegelman BM | title = Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase | journal = The Journal of Biological Chemistry | volume = 271 | issue = 22 | pages = 13018–22 | date = May 1996 | pmid = 8662983 | doi = 10.1074/jbc.271.22.13018 }}</ref> and it is believed that MAP4K4 functions as an upstream signaling element in the TNF-α signaling cascade.<ref name="Yao_1999" />


A recent siRNA screen identified the involvement of MAP4K4 in the regulation of the glucose transporter GLUT4.<ref name="Tang_2006">{{cite journal | vauthors = Tang X, Guilherme A, Chakladar A, Powelka AM, Konda S, Virbasius JV, Nicoloro SM, Straubhaar J, Czech MP | title = An RNA interference-based screen identifies MAP4K4/NIK as a negative regulator of PPARgamma, adipogenesis, and insulin-responsive hexose transport | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 103 | issue = 7 | pages = 2087–92 | date = February 2006 | pmid = 16461467 | doi = 10.1073/pnas.0507660103 }}</ref> The silencing of MAP4K4 in adipocytes elevated the expression of [[Peroxisome proliferator-activated receptor gamma|peroxisome proliferator-activated receptor y]] (PPARy) – a nuclear hormone receptor responsible for the regulation of genes associated with adipocyte differentiation, including [[GLUT4]].<ref>{{cite journal | vauthors = Farmer SR | title = Transcriptional control of adipocyte formation | journal = Cell Metabolism | volume = 4 | issue = 4 | pages = 263–73 | date = October 2006 | pmid = 17011499 | doi = 10.1016/j.cmet.2006.07.001 }}</ref> siRNA silencing of MAP4K4  appears to prevent insulin resistance, restoring insulin sensitivity in human skeletal muscles by down-regulating the TNF-α signaling cascade<ref>{{cite journal | vauthors = Bouzakri K, Zierath JR | title = MAP4K4 gene silencing in human skeletal muscle prevents tumor necrosis factor-alpha-induced insulin resistance | journal = The Journal of Biological Chemistry | volume = 282 | issue = 11 | pages = 7783–9 | date = March 2007 | pmid = 17227768 | doi = 10.1074/jbc.m608602200 }}</ref> and inhibits the TNF-α-induced depletion of PPARy and GLUT4.<ref name="Tang_2006" /> Additionally, miRNA silencing of MAP4K4 in pancreatic beta-cells conferred protection against TNF-α repression of insulin transcription and secretion,<ref name="Zhao_2012">{{cite journal | vauthors = Zhao X, Mohan R, Özcan S, Tang X | title = MicroRNA-30d induces insulin transcription factor MafA and insulin production by targeting mitogen-activated protein 4 kinase 4 (MAP4K4) in pancreatic β-cells | journal = The Journal of Biological Chemistry | volume = 287 | issue = 37 | pages = 31155–64 | date = September 2012 | pmid = 22733810 | doi = 10.1074/jbc.m112.362632 }}</ref> further confirming that MAP4K4 targeting is a potential strategy for diabetes prevention and treatment.<ref name="Zhao_2012" />
A recent siRNA screen identified the involvement of MAP4K4 in the regulation of the glucose transporter GLUT4.<ref name="Tang_2006">{{cite journal | vauthors = Tang X, Guilherme A, Chakladar A, Powelka AM, Konda S, Virbasius JV, Nicoloro SM, Straubhaar J, Czech MP | title = An RNA interference-based screen identifies MAP4K4/NIK as a negative regulator of PPARgamma, adipogenesis, and insulin-responsive hexose transport | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 103 | issue = 7 | pages = 2087–92 | date = February 2006 | pmid = 16461467 | doi = 10.1073/pnas.0507660103 | pmc=1413698}}</ref> The silencing of MAP4K4 in adipocytes elevated the expression of [[Peroxisome proliferator-activated receptor gamma|peroxisome proliferator-activated receptor y]] (PPARy) – a nuclear hormone receptor responsible for the regulation of genes associated with adipocyte differentiation, including [[GLUT4]].<ref>{{cite journal | vauthors = Farmer SR | title = Transcriptional control of adipocyte formation | journal = Cell Metabolism | volume = 4 | issue = 4 | pages = 263–73 | date = October 2006 | pmid = 17011499 | doi = 10.1016/j.cmet.2006.07.001 | pmc=1958996}}</ref> siRNA silencing of MAP4K4  appears to prevent insulin resistance, restoring insulin sensitivity in human skeletal muscles by down-regulating the TNF-α signaling cascade<ref>{{cite journal | vauthors = Bouzakri K, Zierath JR | title = MAP4K4 gene silencing in human skeletal muscle prevents tumor necrosis factor-alpha-induced insulin resistance | journal = The Journal of Biological Chemistry | volume = 282 | issue = 11 | pages = 7783–9 | date = March 2007 | pmid = 17227768 | doi = 10.1074/jbc.m608602200 }}</ref> and inhibits the TNF-α-induced depletion of PPARy and GLUT4.<ref name="Tang_2006" /> Additionally, miRNA silencing of MAP4K4 in pancreatic beta-cells conferred protection against TNF-α repression of insulin transcription and secretion,<ref name="Zhao_2012">{{cite journal | vauthors = Zhao X, Mohan R, Özcan S, Tang X | title = MicroRNA-30d induces insulin transcription factor MafA and insulin production by targeting mitogen-activated protein 4 kinase 4 (MAP4K4) in pancreatic β-cells | journal = The Journal of Biological Chemistry | volume = 287 | issue = 37 | pages = 31155–64 | date = September 2012 | pmid = 22733810 | doi = 10.1074/jbc.m112.362632 | pmc=3438947}}</ref> further confirming that MAP4K4 targeting is a potential strategy for diabetes prevention and treatment.<ref name="Zhao_2012" />


=== Atherosclerosis ===
=== Atherosclerosis ===
[[Atherosclerosis]] is the result of an inflammatory response to lipid-mediated vascular damage. It has been identified that cytokines such as TNF-α induce the expression of pro-inflammatory genes to synthesize leukocyte [[adhesion molecules]] and [[chemokine]]s.<ref>{{cite journal | vauthors = Pober JS | title = Endothelial activation: intracellular signaling pathways | journal = Arthritis Research | volume = 4 Suppl 3 | issue = 3 | pages = S109-16 | date = 2002-05-09 | pmid = 12110129 | doi = 10.1186/ar576 }}</ref> Endothelial cells highly express MAP4K4<ref name="Virbasius_2016" /> and recent studies have reported that MAP4K4 enhances endothelial permeability.<ref>{{cite journal | vauthors = Pannekoek WJ, Linnemann JR, Brouwer PM, Bos JL, Rehmann H | title = Rap1 and Rap2 antagonistically control endothelial barrier resistance | journal = PloS One | volume = 8 | issue = 2 | pages = e57903 | date = 2013-02-28 | pmid = 23469100 | doi = 10.1371/journal.pone.0057903 }}</ref> This consequently contributes to the development of atherosclerosis due to the promotion of [[leukocyte extravasation]], transport of oxidized lipids and the formation of plaques.<ref name="Virbasius_2016" />
[[Atherosclerosis]] is the result of an inflammatory response to lipid-mediated vascular damage. It has been identified that cytokines such as TNF-α induce the expression of pro-inflammatory genes to synthesize leukocyte [[adhesion molecules]] and [[chemokine]]s.<ref>{{cite journal | vauthors = Pober JS | title = Endothelial activation: intracellular signaling pathways | journal = Arthritis Research | volume = 4 Suppl 3 | issue = 3 | pages = S109-16 | date = 2002-05-09 | pmid = 12110129 | doi = 10.1186/ar576 | pmc=3240152}}</ref> Endothelial cells highly express MAP4K4<ref name="Virbasius_2016" /> and recent studies have reported that MAP4K4 enhances endothelial permeability.<ref>{{cite journal | vauthors = Pannekoek WJ, Linnemann JR, Brouwer PM, Bos JL, Rehmann H | title = Rap1 and Rap2 antagonistically control endothelial barrier resistance | journal = PLOS One | volume = 8 | issue = 2 | pages = e57903 | date = 2013-02-28 | pmid = 23469100 | doi = 10.1371/journal.pone.0057903 | pmc=3585282}}</ref> This consequently contributes to the development of atherosclerosis due to the promotion of [[leukocyte extravasation]], transport of oxidized lipids and the formation of plaques.<ref name="Virbasius_2016" />


Silencing of endothelial MAP4K4 ameliorated the development of atherosclerosis in mice.<ref name="Roth_Flach_2015">{{cite journal | vauthors = Roth Flach RJ, Skoura A, Matevossian A, Danai LV, Zheng W, Cortes C, Bhattacharya SK, Aouadi M, Hagan N, Yawe JC, Vangala P, Menendez LG, Cooper MP, Fitzgibbons TP, Buckbinder L, Czech MP | title = Endothelial protein kinase MAP4K4 promotes vascular inflammation and atherosclerosis | journal = Nature Communications | volume = 6 | pages = 8995 | date = December 2015 | pmid = 26688060 | doi = 10.1038/ncomms9995 }}</ref> Additionally, treatment of a MAP4K4 protein kinase inhibitor in mice significantly reduced plaque progression and promoted plaque regression<ref name="Roth_Flach_2015" /> suggesting therapeutic targeting of MAP4K4 may be a beneficial strategy for cardiovascular disease.
Silencing of endothelial MAP4K4 ameliorated the development of atherosclerosis in mice.<ref name="Roth_Flach_2015">{{cite journal | vauthors = Roth Flach RJ, Skoura A, Matevossian A, Danai LV, Zheng W, Cortes C, Bhattacharya SK, Aouadi M, Hagan N, Yawe JC, Vangala P, Menendez LG, Cooper MP, Fitzgibbons TP, Buckbinder L, Czech MP | title = Endothelial protein kinase MAP4K4 promotes vascular inflammation and atherosclerosis | journal = Nature Communications | volume = 6 | pages = 8995 | date = December 2015 | pmid = 26688060 | doi = 10.1038/ncomms9995 | pmc=4703891}}</ref> Additionally, treatment of a MAP4K4 protein kinase inhibitor in mice significantly reduced plaque progression and promoted plaque regression<ref name="Roth_Flach_2015" /> suggesting therapeutic targeting of MAP4K4 may be a beneficial strategy for cardiovascular disease.


=== Cancer ===
=== Cancer ===
The biggest causes of death for patients with cancer are tumour invasion and [[metastasis]] – processes that are highly correlated with cell migration and motility.<ref name="Collins_2006">{{cite journal | vauthors = Collins CS, Hong J, Sapinoso L, Zhou Y, Liu Z, Micklash K, Schultz PG, Hampton GM | title = A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 103 | issue = 10 | pages = 3775–80 | date = March 2006 | pmid = 16537454 | doi = 10.1073/pnas.0600040103 }}</ref> There is limited information regarding how MAP4K4 is involved in cancer but studies have shown that MAP4K4 is overexpressed in a number of cancer types including lung, prostate, pancreatic and ovarian cancer where such up-regulation is associated with increased cell migration, adhesion and invasiveness.<ref name="Buburuzan_2011" />
The biggest causes of death for patients with cancer are tumour invasion and [[metastasis]] – processes that are highly correlated with cell migration and motility.<ref name="Collins_2006">{{cite journal | vauthors = Collins CS, Hong J, Sapinoso L, Zhou Y, Liu Z, Micklash K, Schultz PG, Hampton GM | title = A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 103 | issue = 10 | pages = 3775–80 | date = March 2006 | pmid = 16537454 | doi = 10.1073/pnas.0600040103 | pmc=1383649}}</ref> There is limited information regarding how MAP4K4 is involved in cancer but studies have shown that MAP4K4 is overexpressed in a number of cancer types including lung, prostate, pancreatic and ovarian cancer where such up-regulation is associated with increased cell migration, adhesion and invasiveness.<ref name="Buburuzan_2011" />


Several studies have identified MAP4K4 as an upstream regulator of proteins associated with cytoskeletal dynamics or adhesion. Deletion of the MAP4K4 gene appears to affect membrane dynamics in endothelial cells, resulting in reduced cell migration and impaired angiogenesis;<ref>{{cite journal | vauthors = Vitorino P, Yeung S, Crow A, Bakke J, Smyczek T, West K, McNamara E, Eastham-Anderson J, Gould S, Harris SF, Ndubaku C, Ye W | title = MAP4K4 regulates integrin-FERM binding to control endothelial cell motility | journal = Nature | volume = 519 | issue = 7544 | pages = 425–30 | date = March 2015 | pmid = 25799996 | doi = 10.1038/nature14323 }}</ref> while an overexpression significantly elevates the rate of cell invasion and morphogenesis.<ref name="Wright_2003"/>
Several studies have identified MAP4K4 as an upstream regulator of proteins associated with cytoskeletal dynamics or adhesion. Deletion of the MAP4K4 gene appears to affect membrane dynamics in endothelial cells, resulting in reduced cell migration and impaired angiogenesis;<ref>{{cite journal | vauthors = Vitorino P, Yeung S, Crow A, Bakke J, Smyczek T, West K, McNamara E, Eastham-Anderson J, Gould S, Harris SF, Ndubaku C, Ye W | title = MAP4K4 regulates integrin-FERM binding to control endothelial cell motility | journal = Nature | volume = 519 | issue = 7544 | pages = 425–30 | date = March 2015 | pmid = 25799996 | doi = 10.1038/nature14323 }}</ref> while an overexpression significantly elevates the rate of cell invasion and morphogenesis.<ref name="Wright_2003"/>
Line 68: Line 68:
* {{cite journal | vauthors = Poinat P, De Arcangelis A, Sookhareea S, Zhu X, Hedgecock EM, Labouesse M, Georges-Labouesse E | title = A conserved interaction between beta1 integrin/PAT-3 and Nck-interacting kinase/MIG-15 that mediates commissural axon navigation in C. elegans | journal = Current Biology | volume = 12 | issue = 8 | pages = 622–31 | date = April 2002 | pmid = 11967148 | doi = 10.1016/S0960-9822(02)00764-9 }}
* {{cite journal | vauthors = Poinat P, De Arcangelis A, Sookhareea S, Zhu X, Hedgecock EM, Labouesse M, Georges-Labouesse E | title = A conserved interaction between beta1 integrin/PAT-3 and Nck-interacting kinase/MIG-15 that mediates commissural axon navigation in C. elegans | journal = Current Biology | volume = 12 | issue = 8 | pages = 622–31 | date = April 2002 | pmid = 11967148 | doi = 10.1016/S0960-9822(02)00764-9 }}
* {{cite journal | vauthors = Fong A, Zhang M, Neely J, Sun SC | title = S9, a 19 S proteasome subunit interacting with ubiquitinated NF-kappaB2/p100 | journal = The Journal of Biological Chemistry | volume = 277 | issue = 43 | pages = 40697–702 | date = October 2002 | pmid = 12185077 | doi = 10.1074/jbc.M205330200 }}
* {{cite journal | vauthors = Fong A, Zhang M, Neely J, Sun SC | title = S9, a 19 S proteasome subunit interacting with ubiquitinated NF-kappaB2/p100 | journal = The Journal of Biological Chemistry | volume = 277 | issue = 43 | pages = 40697–702 | date = October 2002 | pmid = 12185077 | doi = 10.1074/jbc.M205330200 }}
* {{cite journal | vauthors = Luan Z, Zhang Y, Liu A, Man Y, Cheng L, Hu G | title = A novel GTP-binding protein hGBP3 interacts with NIK/HGK | journal = FEBS Letters | volume = 530 | issue = 1-3 | pages = 233–8 | date = October 2002 | pmid = 12387898 | doi = 10.1016/S0014-5793(02)03467-1 }}
* {{cite journal | vauthors = Luan Z, Zhang Y, Liu A, Man Y, Cheng L, Hu G | title = A novel GTP-binding protein hGBP3 interacts with NIK/HGK | journal = FEBS Letters | volume = 530 | issue = 1–3 | pages = 233–8 | date = October 2002 | pmid = 12387898 | doi = 10.1016/S0014-5793(02)03467-1 }}
* {{cite journal | vauthors = Wright JH, Wang X, Manning G, LaMere BJ, Le P, Zhu S, Khatry D, Flanagan PM, Buckley SD, Whyte DB, Howlett AR, Bischoff JR, Lipson KE, Jallal B | title = The STE20 kinase HGK is broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion | journal = Molecular and Cellular Biology | volume = 23 | issue = 6 | pages = 2068–82 | date = March 2003 | pmid = 12612079 | pmc = 149462 | doi = 10.1128/MCB.23.6.2068-2082.2003 }}
* {{cite journal | vauthors = Wright JH, Wang X, Manning G, LaMere BJ, Le P, Zhu S, Khatry D, Flanagan PM, Buckley SD, Whyte DB, Howlett AR, Bischoff JR, Lipson KE, Jallal B | title = The STE20 kinase HGK is broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion | journal = Molecular and Cellular Biology | volume = 23 | issue = 6 | pages = 2068–82 | date = March 2003 | pmid = 12612079 | pmc = 149462 | doi = 10.1128/MCB.23.6.2068-2082.2003 }}
* {{cite journal | vauthors = Rodriguez M, Yu X, Chen J, Songyang Z | title = Phosphopeptide binding specificities of BRCA1 COOH-terminal (BRCT) domains | journal = The Journal of Biological Chemistry | volume = 278 | issue = 52 | pages = 52914–8 | date = December 2003 | pmid = 14578343 | doi = 10.1074/jbc.C300407200 }}
* {{cite journal | vauthors = Rodriguez M, Yu X, Chen J, Songyang Z | title = Phosphopeptide binding specificities of BRCA1 COOH-terminal (BRCT) domains | journal = The Journal of Biological Chemistry | volume = 278 | issue = 52 | pages = 52914–8 | date = December 2003 | pmid = 14578343 | doi = 10.1074/jbc.C300407200 }}

Latest revision as of 08:37, 23 March 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

Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) – also known as hepatocyte progenitor kinase-like/germinal center kinase-like kinase (HGK) and Nck-interacting kinase (NIK) – is an enzyme, specifically a serine/threonine (S/T) kinase encoded by the MAP4K4 gene in humans.[1][2]

MAP4K4 is involved in a wide array of physiological processes including cell migration, proliferation and adhesion;[3] its activity has been implicated in systemic inflammation,[4] metabolic disorders,[5] cardiovascular disease[5] and cancer.[2]

While MAP4K4 has been found to be upregulated in a wide array of cancers, there is currently limited information regarding its specific involvement. However, there is increasing evidence that suggests MAP4K4 has an important role in the development and progression of cancer, and may serve as a novel target for cancer therapeutics.[2]

Discovery and classification

MAP4K4 is categorized under the mammalian sterile 20 protein (Ste20p) kinase family due to its shared homology with the Ste20p kinase found in budding yeast[1] and is a member of the GCK-IV subfamily. Mammalian MAP4K4 was initially identified in mice as a kinase activator for a protein called Nck[6] followed shortly by identification and cloning of the human orthologue encoded by the MAP4K4 gene.[7]

Structure and expression

In humans, MAP4K4 is encoded by the MAP4K4 gene located on chromosome 2, position q11.2 and consists of 33 exons responsible for its synthesis.[1] It contains approximately 1200 amino acids, has a molecular mass of ~140 KDa.[6][7] and its orthologues across various species share molecular and structural similarities.

Structurally MAP4K4 contains the following domains:[1]

  • N-Terminal Kinase Domain
  • Coiled-coil domain
  • C-Terminal Hydrophobic Leucine-Rich Citron Homology Domain (CNH)
  • Interdomain - Connects the kinase and CNH domains, facilitates protein-protein interactions. Although it has been identified, its structural components and functionality are currently poorly understood
File:MAP4K4 V2.jpg
Figure 1. Schematic representation of MAP4K4 structure, depicts the N-terminal kinase domain, C-terminal citron homology domain (regulatory function) and the interdomain (facilitates protein interactions).[2][8] Note: the numbers indicate amino acid positions.

Alternative splicing of the MAP4K4 gene yields five functional isoforms, all of which display fully homologous kinase and CNH domains but differences in the interdomain domain.[9] While the biological significance of these isoforms remains to be determined, it can be speculated that such variations alter and determine MAP4K4's interactions with other proteins and factors, ultimately leading to the activation/inhibition of different biochemical and physiological cascades.

The mammalian class of Ste20 kinases require phosphorylation at specific sites for full activity. Primary phosphorylation at the activation site in their kinase domain is believed to cause a conformational change in the protein, stabilizing the structure of its activation segment to allow suitable substrate binding.[1] Secondary sites also require phosphorylation for the enzyme to assume full activation and is achieved via autophosphorylation or by upstream kinases.[1]

To date, MAP4K4 has been found to be expressed in all tissue types[7] with a relatively more pronounced expression in the brain and testes.[10] Multiple isoforms of MAP4K4 can be present at any given time in the same cell but the abundance of each isoform in the cell differs depending on the cell-type or tissue-type.[10]

  • E.g. In humans, the shorter isoform of MAP4K4 is predominantly expressed in organs including the liver, placenta, skeletal muscles while a longer isoform is expressed in the brain

Interactions and signaling

TNF-α

Evidence from mammalian and fly studies indicate that MAP4K4 is involved with tumour necrosis factor alpha (TNF-α) and its c-jun N-terminal kinase (JNK) signaling pathway.[11] MAP4K4 not only mediates TNF-α signaling but also promotes its expression;[7] moreover, TNF-α can elevate MAP4K4 expression using transcription factors[12]

The JNK pathway is implicated in a number of physiological processes and involves JNKs – kinases responsible for the phosphorylation of a downstream protein called c-Jun. This further leads to the increase in expression and activity of specific transcription factors that respond to a variety of cellular stressors, growth factors and cytokines. The activation of the JNK signal transduction pathway by MAP4K4 has been implicated in apoptotic regulation of many different cell types,[13] tumorigenesis and/or inflammation.[3]

p53

p53 is a tumour suppressor gene and is involved with cellular response to stress. When expressed, the cell cycle is halted in the G1 phase and can induce senescence or apoptosis. Mutations to the p53 gene are often found in many types of cancers.

The MAP4K4 gene contains four binding sites for p53. Upon binding, p53 up-regulates MAP4K4 expression leading to the activation of the JNK signaling pathway. siRNA knockdown experiments have also shown a reduction of p53 induced apoptosis.[13] Current evidence therefore suggests that MAP4K4 has a modulating effect on p53 induced apoptosis in the JNK signaling pathway.

Clinical significance

Glucose uptake and insulin function

MAP4K4 has been identified to be involved in the negative regulation of insulin-dependent glucose transport. There is increasing evidence suggesting cytokines such as TNF-α mediate biological effects antagonistic to insulin action and induce inflammation observed in obesity.[14][15] TNF-α specifically attenuates the signaling pathway initiated by insulin receptors, reducing the amount of glucose transport and uptake;[16] and it is believed that MAP4K4 functions as an upstream signaling element in the TNF-α signaling cascade.[7]

A recent siRNA screen identified the involvement of MAP4K4 in the regulation of the glucose transporter GLUT4.[17] The silencing of MAP4K4 in adipocytes elevated the expression of peroxisome proliferator-activated receptor y (PPARy) – a nuclear hormone receptor responsible for the regulation of genes associated with adipocyte differentiation, including GLUT4.[18] siRNA silencing of MAP4K4 appears to prevent insulin resistance, restoring insulin sensitivity in human skeletal muscles by down-regulating the TNF-α signaling cascade[19] and inhibits the TNF-α-induced depletion of PPARy and GLUT4.[17] Additionally, miRNA silencing of MAP4K4 in pancreatic beta-cells conferred protection against TNF-α repression of insulin transcription and secretion,[20] further confirming that MAP4K4 targeting is a potential strategy for diabetes prevention and treatment.[20]

Atherosclerosis

Atherosclerosis is the result of an inflammatory response to lipid-mediated vascular damage. It has been identified that cytokines such as TNF-α induce the expression of pro-inflammatory genes to synthesize leukocyte adhesion molecules and chemokines.[21] Endothelial cells highly express MAP4K4[5] and recent studies have reported that MAP4K4 enhances endothelial permeability.[22] This consequently contributes to the development of atherosclerosis due to the promotion of leukocyte extravasation, transport of oxidized lipids and the formation of plaques.[5]

Silencing of endothelial MAP4K4 ameliorated the development of atherosclerosis in mice.[23] Additionally, treatment of a MAP4K4 protein kinase inhibitor in mice significantly reduced plaque progression and promoted plaque regression[23] suggesting therapeutic targeting of MAP4K4 may be a beneficial strategy for cardiovascular disease.

Cancer

The biggest causes of death for patients with cancer are tumour invasion and metastasis – processes that are highly correlated with cell migration and motility.[24] There is limited information regarding how MAP4K4 is involved in cancer but studies have shown that MAP4K4 is overexpressed in a number of cancer types including lung, prostate, pancreatic and ovarian cancer where such up-regulation is associated with increased cell migration, adhesion and invasiveness.[3]

Several studies have identified MAP4K4 as an upstream regulator of proteins associated with cytoskeletal dynamics or adhesion. Deletion of the MAP4K4 gene appears to affect membrane dynamics in endothelial cells, resulting in reduced cell migration and impaired angiogenesis;[25] while an overexpression significantly elevates the rate of cell invasion and morphogenesis.[10]

Evidence also indicates that MAP4K4 is a major contributor to the elevated growth and migratory properties of tumour cells.[24][26] Poor prognosis and clinical progression of hepatocellular carcinoma,[26] pancreatic adenocarcinoma,[27] and colorectal cancer[28] are all closely correlated with MAP4K4 expression levels.

References

  1. 1.0 1.1 1.2 1.3 1.4 1.5 Delpire E (September 2009). "The mammalian family of sterile 20p-like protein kinases". Pflügers Archiv. 458 (5): 953–67. doi:10.1007/s00424-009-0674-y. PMID 19399514.
  2. 2.0 2.1 2.2 2.3 Gao X, Gao C, Liu G, Hu J (2016-10-28). "MAP4K4: an emerging therapeutic target in cancer". Cell & Bioscience. 6: 56. doi:10.1186/s13578-016-0121-7. PMC 5084373. PMID 27800153.
  3. 3.0 3.1 3.2 Buburuzan L, Luca C (2011). "MAP4K4 a possible new biomarker in cancer therapy". Analele Stiintifice ale Universitatii" Al. I. Cuza" Din Iasi.(Serie Noua). Sectiunea 2. a. Genetica si Biologie Moleculara. Universitatea" Alexandru Ioan Cuza". 12 (2).
  4. Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP (April 2009). "Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation". Nature. 458 (7242): 1180–4. doi:10.1038/nature07774. PMC 2879154. PMID 19407801.
  5. 5.0 5.1 5.2 5.3 Virbasius JV, Czech MP (July 2016). "Map4k4 Signaling Nodes in Metabolic and Cardiovascular Diseases". Trends in Endocrinology and Metabolism. 27 (7): 484–92. doi:10.1016/j.tem.2016.04.006. PMC 4912878. PMID 27160798.
  6. 6.0 6.1 Su YC, Han J, Xu S, Cobb M, Skolnik EY (March 1997). "NIK is a new Ste20-related kinase that binds NCK and MEKK1 and activates the SAPK/JNK cascade via a conserved regulatory domain". The EMBO Journal. 16 (6): 1279–90. doi:10.1093/emboj/16.6.1279. PMC 1169726. PMID 9135144.
  7. 7.0 7.1 7.2 7.3 7.4 Yao Z, Zhou G, Wang XS, Brown A, Diener K, Gan H, Tan TH (January 1999). "A novel human STE20-related protein kinase, HGK, that specifically activates the c-Jun N-terminal kinase signaling pathway". The Journal of Biological Chemistry. 274 (4): 2118–25. doi:10.1074/jbc.274.4.2118. PMID 9890973.
  8. "MAP4K4 - Mitogen-activated protein kinase kinase kinase kinase 4 - Homo sapiens (Human) - MAP4K4 gene & protein". www.uniprot.org. Retrieved 2017-11-30.
  9. Santhana Kumar K, Tripolitsioti D, Ma M, Grählert J, Egli KB, Fiaschetti G, Shalaby T, Grotzer MA, Baumgartner M (2015). "The Ser/Thr kinase MAP4K4 drives c-Met-induced motility and invasiveness in a cell-based model of SHH medulloblastoma". SpringerPlus. 4: 19. doi:10.1186/s40064-015-0784-2. PMC 4302160. PMID 25625039.
  10. 10.0 10.1 10.2 Wright JH, Wang X, Manning G, LaMere BJ, Le P, Zhu S, Khatry D, Flanagan PM, Buckley SD, Whyte DB, Howlett AR, Bischoff JR, Lipson KE, Jallal B (March 2003). "The STE20 kinase HGK is broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion". Molecular and Cellular Biology. 23 (6): 2068–82. doi:10.1128/mcb.23.6.2068-2082.2003. PMC 149462. PMID 12612079.
  11. Liu H, Su YC, Becker E, Treisman J, Skolnik EY (1999). "A Drosophila TNF-receptor-associated factor (TRAF) binds the ste20 kinase Misshapen and activates Jun kinase". Current Biology. 9 (2): 101–4. doi:10.1016/s0960-9822(99)80023-2. PMID 10021364.
  12. Tesz GJ, Guilherme A, Guntur KV, Hubbard AC, Tang X, Chawla A, Czech MP (July 2007). "Tumor necrosis factor alpha (TNFalpha) stimulates Map4k4 expression through TNFalpha receptor 1 signaling to c-Jun and activating transcription factor 2". The Journal of Biological Chemistry. 282 (27): 19302–12. doi:10.1074/jbc.m700665200. PMID 17500068.
  13. 13.0 13.1 Miled C, Pontoglio M, Garbay S, Yaniv M, Weitzman JB (June 2005). "A genomic map of p53 binding sites identifies novel p53 targets involved in an apoptotic network". Cancer Research. 65 (12): 5096–104. doi:10.1158/0008-5472.can-04-4232. PMID 15958553.
  14. Hotamisligil GS, Spiegelman BM (November 1994). "Tumor necrosis factor alpha: a key component of the obesity-diabetes link". Diabetes. 43 (11): 1271–8. doi:10.2337/diab.43.11.1271. PMID 7926300.
  15. Skolnik EY, Marcusohn J (1996). "Inhibition of insulin receptor signaling by TNF: potential role in obesity and non-insulin-dependent diabetes mellitus". Cytokine & Growth Factor Reviews. 7 (2): 161–73. doi:10.1016/1359-6101(96)00021-4. PMID 8899294.
  16. Peraldi P, Hotamisligil GS, Buurman WA, White MF, Spiegelman BM (May 1996). "Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase". The Journal of Biological Chemistry. 271 (22): 13018–22. doi:10.1074/jbc.271.22.13018. PMID 8662983.
  17. 17.0 17.1 Tang X, Guilherme A, Chakladar A, Powelka AM, Konda S, Virbasius JV, Nicoloro SM, Straubhaar J, Czech MP (February 2006). "An RNA interference-based screen identifies MAP4K4/NIK as a negative regulator of PPARgamma, adipogenesis, and insulin-responsive hexose transport". Proceedings of the National Academy of Sciences of the United States of America. 103 (7): 2087–92. doi:10.1073/pnas.0507660103. PMC 1413698. PMID 16461467.
  18. Farmer SR (October 2006). "Transcriptional control of adipocyte formation". Cell Metabolism. 4 (4): 263–73. doi:10.1016/j.cmet.2006.07.001. PMC 1958996. PMID 17011499.
  19. Bouzakri K, Zierath JR (March 2007). "MAP4K4 gene silencing in human skeletal muscle prevents tumor necrosis factor-alpha-induced insulin resistance". The Journal of Biological Chemistry. 282 (11): 7783–9. doi:10.1074/jbc.m608602200. PMID 17227768.
  20. 20.0 20.1 Zhao X, Mohan R, Özcan S, Tang X (September 2012). "MicroRNA-30d induces insulin transcription factor MafA and insulin production by targeting mitogen-activated protein 4 kinase 4 (MAP4K4) in pancreatic β-cells". The Journal of Biological Chemistry. 287 (37): 31155–64. doi:10.1074/jbc.m112.362632. PMC 3438947. PMID 22733810.
  21. Pober JS (2002-05-09). "Endothelial activation: intracellular signaling pathways". Arthritis Research. 4 Suppl 3 (3): S109–16. doi:10.1186/ar576. PMC 3240152. PMID 12110129.
  22. Pannekoek WJ, Linnemann JR, Brouwer PM, Bos JL, Rehmann H (2013-02-28). "Rap1 and Rap2 antagonistically control endothelial barrier resistance". PLOS One. 8 (2): e57903. doi:10.1371/journal.pone.0057903. PMC 3585282. PMID 23469100.
  23. 23.0 23.1 Roth Flach RJ, Skoura A, Matevossian A, Danai LV, Zheng W, Cortes C, Bhattacharya SK, Aouadi M, Hagan N, Yawe JC, Vangala P, Menendez LG, Cooper MP, Fitzgibbons TP, Buckbinder L, Czech MP (December 2015). "Endothelial protein kinase MAP4K4 promotes vascular inflammation and atherosclerosis". Nature Communications. 6: 8995. doi:10.1038/ncomms9995. PMC 4703891. PMID 26688060.
  24. 24.0 24.1 Collins CS, Hong J, Sapinoso L, Zhou Y, Liu Z, Micklash K, Schultz PG, Hampton GM (March 2006). "A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase". Proceedings of the National Academy of Sciences of the United States of America. 103 (10): 3775–80. doi:10.1073/pnas.0600040103. PMC 1383649. PMID 16537454.
  25. Vitorino P, Yeung S, Crow A, Bakke J, Smyczek T, West K, McNamara E, Eastham-Anderson J, Gould S, Harris SF, Ndubaku C, Ye W (March 2015). "MAP4K4 regulates integrin-FERM binding to control endothelial cell motility". Nature. 519 (7544): 425–30. doi:10.1038/nature14323. PMID 25799996.
  26. 26.0 26.1 Liu AW, Cai J, Zhao XL, Jiang TH, He TF, Fu HQ, Zhu MH, Zhang SH (February 2011). "ShRNA-targeted MAP4K4 inhibits hepatocellular carcinoma growth". Clinical Cancer Research. 17 (4): 710–20. doi:10.1158/1078-0432.ccr-10-0331. PMID 21196414.
  27. Liang JJ, Wang H, Rashid A, Tan TH, Hwang RF, Hamilton SR, Abbruzzese JL, Evans DB, Wang H (November 2008). "Expression of MAP4K4 is associated with worse prognosis in patients with stage II pancreatic ductal adenocarcinoma". Clinical Cancer Research. 14 (21): 7043–9. doi:10.1158/1078-0432.ccr-08-0381. PMID 18981001.
  28. Hao JM, Chen JZ, Sui HM, Si-Ma XQ, Li GQ, Liu C, Li JL, Ding YQ, Li JM (March 2010). "A five-gene signature as a potential predictor of metastasis and survival in colorectal cancer". The Journal of Pathology. 220 (4): 475–89. doi:10.1002/path.2668. PMID 20077526.

Further reading