C-Fos: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

In the fields of molecular biology and genetics, c-fos is a proto-oncogene that is the human homolog of the retroviral oncogene v-fos. It was first discovered in rat fibroblasts as the transforming gene of the FBJ MSV (Finkel–Biskis–Jinkins murine osteogenic sarcoma virus) (Curran and Tech, 1982). It is a part of a bigger Fos family of transcription factors which includes c-Fos, FosB, Fra-1 and Fra-2. It has been mapped to chromosome region 14q21→q31. c-Fos encodes a 62 kDa protein, which forms heterodimer with c-jun (part of Jun family of transcription factors), resulting in the formation of AP-1 (Activator Protein-1) complex which binds DNA at AP-1 specific sites at the promoter and enhancer regions of target genes and converts extracellular signals into changes of gene expression. It plays an important role in many cellular functions and has been found to be overexpressed in a variety of cancers.

  • cellular functions
  • molecular biology
  • retroviral

1. Structure and Function

c-Fos is a 380 amino acid protein with a basic leucine zipper region for dimerisation and DNA-binding and a transactivation domain at C-terminus, and, like Jun proteins, it can form homodimers.[1] In vitro studies have shown that Jun–Fos heterodimers are more stable and have stronger DNA-binding activity than Jun–Jun homodimers.[2]

A variety of stimuli, including serum, growth factors, tumor promoters, cytokines, and UV radiation induce their expression. The c-fos mRNA and protein is generally among the first to be expressed and hence referred to as an immediate early gene. It is rapidly and transiently induced, within 15 minutes of stimulation.[3] Its activity is also regulated by posttranslational modification caused by phosphorylation by different kinases, like MAPK, CDC2, PKA or PKC which influence protein stability, DNA-binding activity and the trans-activating potential of the transcription factors.[4][5][6] It can cause gene repression as well as gene activation, although different domains are believed to be involved in both processes.

It is involved in important cellular events, including cell proliferation, differentiation and survival; genes associated with hypoxia; and angiogenesis;[7] which makes its dysregulation an important factor for cancer development. It can also induce a loss of cell polarity and epithelial-mesenchymal transition, leading to invasive and metastatic growth in mammary epithelial cells.[8]

The importance of c-fos in biological context has been determined by eliminating endogenous function by using anti-sense mRNA, anti-c-fos antibodies, a ribozyme that cleaves c-fos mRNA or a dominant negative mutant of c-fos. The transgenic mice thus generated are viable, demonstrating that there are c-fos dependent and independent pathways of cell proliferation, but display a range of tissue-specific developmental defects, including osteoporosis, delayed gametogenesis, lymphopenia and behavioral abnormalities.

2. Clinical Significance

The AP-1 complex has been implicated in transformation and progression of cancer. In osteosarcoma and endometrial carcinoma, c-Fos overexpression was associated with high-grade lesions and poor prognosis. Also, in a comparison between precancerous lesion of the cervix uteri and invasive cervical cancer, c-Fos expression was significantly lower in precancerous lesions. c-Fos has also been identified as independent predictor of decreased survival in breast cancer.[9]

It was found that overexpression of c-fos from class I MHC promoter in transgenic mice leads to the formation of osteosarcomas due to increased proliferation of osteoblasts whereas ectopic expression of the other Jun and Fos proteins does not induce any malignant tumors. Activation of the c-Fos transgene in mice results in overexpression of cyclin D1, A and E in osteoblasts and chondrocytes, both in vitro and in vivo, which might contribute to the uncontrolled growth leading to tumor. Human osteosarcomas analyzed for c-fos expression have given positive results in more than half the cases and c-fos expression has been associated with higher frequency of relapse and poor response to chemotherapy.

Several studies have raised the idea that c-Fos may also have tumor-suppressor activity, that it might be able to promote as well as suppress tumorigenesis. Supporting this is the observation that in ovarian carcinomas, loss of c-Fos expression correlates with disease progression. This double action could be enabled by differential protein composition of tumour cells and their environment, for example, dimerisation partners, co-activators and promoter architecture. It is possible that the tumor suppressing activity is due to a proapoptotic function. The exact mechanism by which c-Fos contributes to apoptosis is not clearly understood, but observations in human hepatocellular carcinoma cells indicate that c-Fos is a mediator of c-myc-induced cell death and might induce apoptosis through the p38 MAP kinase pathway. Fas ligand (FASLG or FasL) and the tumour necrosis factor-related apoptosis-inducing ligand (TNFSF10 or TRAIL) might reflect an additional apoptotic mechanism induced by c-Fos, as observed in a human T-cell leukaemia cell line. Another possible mechanism of c-Fos involvement in tumour suppression could be the direct regulation of BRCA1, a well established factor in familial breast and ovarian cancer.

In addition, the role of c-fos and other Fos family proteins has also been studied in endometrial carcinoma, cervical cancer, mesotheliomas, colorectal cancer, lung cancer, melanomas, thyroid carcinomas, esophageal cancer, hepatocellular carcinomas, etc.

Cocaine, methamphetamine,[10] morphine,[11] and other psychoactive drugs[12][13] have been shown to increase c-Fos production in the mesocortical pathway (prefrontal cortex) as well as in the mesolimbic reward pathway (nucleus accumbens), as well as display variability depending on prior sensitization.[13] c-Fos repression by ΔFosB's AP-1 complex within the D1-type medium spiny neurons of the nucleus accumbens acts as a molecular switch that enables the chronic induction of ΔFosB, thus allowing it to accumulate more rapidly. As such, the c-Fos promoter finds utilization in drug addiction research in general, as well as with context-induced relapse to drug-seeking and other behavioral changes associated with chronic drug taking.

An increase in c-Fos production in androgen receptor-containing neurons has been observed in rats after mating.

3. Applications

Expression of c-fos is an indirect marker of neuronal activity because c-fos is often expressed when neurons fire action potentials.[14][15] Upregulation of c-fos mRNA in a neuron indicates recent activity.[16]

The c-fos promoter has also been utilised for drug abuse research. Scientists use this promoter to turn on transgenes in rats, allowing them to manipulate specific neuronal ensembles to assess their role in drug-related memories and behavior.[17] This neuronal control can be replicated with optogenetics or DREADDs [18]

4. Interactions

c-Fos has been shown to interact with:

Mixed neural cultures derived from rat embryos were grown under normal conditions (left) or treated with 55mM Potassium for 5 hours (right). Cultures were then stained with antibody to the intermediate filament protein vimentin (green), antibody to cFos (red) and with a DNA binding dye (blue). The vimentin antibody reveals non-neuronal cells and the DNA dye shows the nuclei of all cells. The potassium treatment depolarizes the neurons and induces strong expression of cFos in neuronal cell bodies as shown in the right image. Cell culture, Image and antibody generation all performed in the EnCor Biotechnology laboratory. https://handwiki.org/wiki/index.php?curid=1745657

The content is sourced from: https://handwiki.org/wiki/Biology:C-Fos

References

  1. "Evidence for homodimerization of the c-Fos transcription factor in live cells revealed by fluorescence microscopy and computer modeling". Mol. Cell. Biol. 35 (21): 3785–98. November 2015. doi:10.1128/MCB.00346-15. PMID 26303532.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=4589601
  2. "c-Jun dimerizes with itself and with c-Fos, forming complexes of different DNA binding affinities". Cell 55 (5): 917–24. December 1988. doi:10.1016/0092-8674(88)90147-X. PMID 3142692. http://genesdev.cshlp.org/content/2/12b/1687.full.pdf. 
  3. "Targeted disruption of the c-fos gene demonstrates c-fos-dependent and -independent pathways for gene expression stimulated by growth factors or oncogenes". EMBO J. 13 (13): 3094–103. July 1994. doi:10.1002/j.1460-2075.1994.tb06608.x. PMID 8039503.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=395200
  4. "Regulation of Fra-1 and Fra-2 phosphorylation differs during the cell cycle of fibroblasts and phosphorylation in vitro by MAP kinase affects DNA binding activity". Oncogene 9 (9): 2537–47. September 1994. PMID 8058317.  http://www.ncbi.nlm.nih.gov/pubmed/8058317
  5. "Dual role for mitogen-activated protein kinase (Erk) in insulin-dependent regulation of Fra-1 (fos-related antigen-1) transcription and phosphorylation". Biochem. J. 368 (Pt 2): 573–80. December 2002. doi:10.1042/BJ20020579. PMID 12197835.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=1223008
  6. "Extracellular signal-regulated kinase 1/2-mediated phosphorylation of JunD and FosB is required for okadaic acid-induced activator protein 1 activation". J. Biol. Chem. 274 (2): 1124–30. January 1999. doi:10.1074/jbc.274.2.1124. PMID 9873060.  https://dx.doi.org/10.1074%2Fjbc.274.2.1124
  7. Tulchinsky E (July 2000). "Fos family members: regulation, structure and role in oncogenic transformation". Histol. Histopathol. 15 (3): 921–8. PMID 10963134.  http://www.ncbi.nlm.nih.gov/pubmed/10963134
  8. "The estrogen-dependent c-JunER protein causes a reversible loss of mammary epithelial cell polarity involving a destabilization of adherens junctions". J. Cell Biol. 132 (6): 1115–32. March 1996. doi:10.1083/jcb.132.6.1115. PMID 8601589.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2120757
  9. "C-Fos expression is a molecular predictor of progression and survival in epithelial ovarian carcinoma". Br. J. Cancer 99 (8): 1269–75. October 2008. doi:10.1038/sj.bjc.6604650. PMID 18854825.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2570515
  10. "Amphetamine and cocaine induce drug-specific activation of the c-fos gene in striosome-matrix compartments and limbic subdivisions of the striatum". Proc. Natl. Acad. Sci. U.S.A. 87 (17): 6912–6. September 1990. doi:10.1073/pnas.87.17.6912. PMID 2118661. Bibcode: 1990PNAS...87.6912G.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=54648
  11. "Psychomotor stimulant- and opiate-induced c-fos mRNA expression patterns in the rat forebrain: comparisons between acute drug treatment and a drug challenge in sensitized animals". Neurochem. Res. 21 (11): 1425–35. November 1996. doi:10.1007/BF02532384. PMID 8947933.  https://dx.doi.org/10.1007%2FBF02532384
  12. "A single dose of lysergic acid diethylamide influences gene expression patterns within the mammalian brain". Neuropsychopharmacology 26 (5): 634–42. May 2002. doi:10.1016/S0893-133X(01)00405-5. PMID 11927188.  https://dx.doi.org/10.1016%2FS0893-133X%2801%2900405-5
  13. "Induction of c-Fos expression in specific areas of the fear circuitry in rat forebrain by anxiogenic drugs". Biol. Psychiatry 53 (4): 275–83. February 2003. doi:10.1016/S0006-3223(02)01574-3. PMID 12586446.  https://dx.doi.org/10.1016%2FS0006-3223%2802%2901574-3
  14. "Environmental novelty is associated with a selective increase in c-fos expression in the output elements of the hippocampal formation and the perirhinal cortex". Learn. Mem. 15 (12): 899–908. 2008. doi:10.1101/lm.1196508. PMID 19050162.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2632843
  15. "The use of c-fos as a metabolic marker in neuronal pathway tracing". Journal of Neuroscience Methods 29 (3): 261–265. 1989. doi:10.1016/0165-0270(89)90150-7. PMID 2507830.  https://dx.doi.org/10.1016%2F0165-0270%2889%2990150-7
  16. "Conditioned Fear Inhibits c-fos mRNA Expression in the Central Extended Amygdala". Brain Res. 1229: 137–46. September 2008. doi:10.1016/j.brainres.2008.06.085. PMID 18634767.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2605076
  17. "Targeted disruption of cocaine-activated nucleus accumbens neurons prevents context-specific sensitization". Nat. Neurosci. 12 (8): 1069–73. August 2009. doi:10.1038/nn.2364. PMID 19620976.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2752202
  18. Garner, Aleena (March 2012). "Generation of a Synthetic Memory Trace". Science 335 (6075): 1513–1516. doi:10.1126/science.1214985. PMID 22442487. Bibcode: 2012Sci...335.1513G.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=3956300
  19. "Bcl3, an IkappaB protein, stimulates activating protein-1 transactivation and cellular proliferation". J. Biol. Chem. 274 (40): 28491–6. October 1999. doi:10.1074/jbc.274.40.28491. PMID 10497212.  https://dx.doi.org/10.1074%2Fjbc.274.40.28491
  20. "COBRA1 inhibits AP-1 transcriptional activity in transfected cells". Biochem. Biophys. Res. Commun. 325 (2): 568–73. December 2004. doi:10.1016/j.bbrc.2004.10.079. PMID 15530430.  https://dx.doi.org/10.1016%2Fj.bbrc.2004.10.079
  21. "Casein kinase II interacts with the bZIP domains of several transcription factors". Nucleic Acids Res. 26 (16): 3854–61. August 1998. doi:10.1093/nar/26.16.3854. PMID 9685505.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=147779
  22. "CHOP enhancement of gene transcription by interactions with Jun/Fos AP-1 complex proteins". Mol. Cell. Biol. 19 (11): 7589–99. November 1999. doi:10.1128/MCB.19.11.7589. PMID 10523647.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=84780
  23. "ERK MAP kinase links cytokine signals to activation of latent HIV-1 infection by stimulating a cooperative interaction of AP-1 and NF-kappaB". J. Biol. Chem. 274 (39): 27981–8. September 1999. doi:10.1074/jbc.274.39.27981. PMID 10488148.  https://dx.doi.org/10.1074%2Fjbc.274.39.27981
  24. "Identification of SWI.SNF complex subunit BAF60a as a determinant of the transactivation potential of Fos/Jun dimers". J. Biol. Chem. 276 (4): 2852–7. January 2001. doi:10.1074/jbc.M009633200. PMID 11053448.  https://dx.doi.org/10.1074%2Fjbc.M009633200
  25. "Cross-family interaction between the bHLHZip USF and bZip Fra1 proteins results in down-regulation of AP1 activity". Oncogene 14 (17): 2091–8. May 1997. doi:10.1038/sj.onc.1201046. PMID 9160889.  https://dx.doi.org/10.1038%2Fsj.onc.1201046
  26. "Crystal structure of the heterodimeric bZIP transcription factor c-Fos-c-Jun bound to DNA". Nature 373 (6511): 257–61. January 1995. doi:10.1038/373257a0. PMID 7816143. Bibcode: 1995Natur.373..257G.  https://dx.doi.org/10.1038%2F373257a0
  27. "Isolation and characterization of a novel member of the gene family encoding the cAMP response element-binding protein CRE-BP1". J. Biol. Chem. 268 (6): 4259–66. February 1993. doi:10.1016/S0021-9258(18)53604-8. PMID 8440710.  https://dx.doi.org/10.1016%2FS0021-9258%2818%2953604-8
  28. "Detection and modulation in vivo of helix-loop-helix protein-protein interactions". J. Biol. Chem. 268 (1): 5–8. January 1993. doi:10.1016/S0021-9258(18)54105-3. PMID 8380166.  https://dx.doi.org/10.1016%2FS0021-9258%2818%2954105-3
  29. "Nrf2 and Nrf1 in association with Jun proteins regulate antioxidant response element-mediated expression and coordinated induction of genes encoding detoxifying enzymes". Oncogene 17 (24): 3145–56. December 1998. doi:10.1038/sj.onc.1202237. PMID 9872330.  https://dx.doi.org/10.1038%2Fsj.onc.1202237
  30. "Activating protein-1, nuclear factor-kappaB, and serum response factor as novel target molecules of the cancer-amplified transcription coactivator ASC-2". Mol. Endocrinol. 14 (6): 915–25. June 2000. doi:10.1210/mend.14.6.0471. PMID 10847592.  https://dx.doi.org/10.1210%2Fmend.14.6.0471
  31. "Steroid receptor coactivator-1 coactivates activating protein-1-mediated transactivations through interaction with the c-Jun and c-Fos subunits". J. Biol. Chem. 273 (27): 16651–4. July 1998. doi:10.1074/jbc.273.27.16651. PMID 9642216.  https://dx.doi.org/10.1074%2Fjbc.273.27.16651
  32. "Silencing mediator of retinoic acid and thyroid hormone receptors, as a novel transcriptional corepressor molecule of activating protein-1, nuclear factor-kappaB, and serum response factor". J. Biol. Chem. 275 (17): 12470–4. April 2000. doi:10.1074/jbc.275.17.12470. PMID 10777532.  https://dx.doi.org/10.1074%2Fjbc.275.17.12470
  33. "AP-1 and Cbfa/runt physically interact and regulate parathyroid hormone-dependent MMP13 expression in osteoblasts through a new osteoblast-specific element 2/AP-1 composite element". J. Biol. Chem. 276 (23): 20029–38. June 2001. doi:10.1074/jbc.M010601200. PMID 11274169.  https://dx.doi.org/10.1074%2Fjbc.M010601200
  34. "Physical interaction of the activator protein-1 factors c-Fos and c-Jun with Cbfa1 for collagenase-3 promoter activation". J. Biol. Chem. 277 (1): 816–22. January 2002. doi:10.1074/jbc.M107082200. PMID 11641401.  https://dx.doi.org/10.1074%2Fjbc.M107082200
  35. "Smad3 and Smad4 cooperate with c-Jun/c-Fos to mediate TGF-beta-induced transcription". Nature 394 (6696): 909–13. August 1998. doi:10.1038/29814. PMID 9732876. Bibcode: 1998Natur.394..909Z.  https://dx.doi.org/10.1038%2F29814
  36. "c-Fos-induced activation of a TATA-box-containing promoter involves direct contact with TATA-box-binding protein". Mol. Cell. Biol. 14 (9): 6021–9. September 1994. doi:10.1128/MCB.14.9.6021. PMID 8065335.  http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=359128
More
This entry is offline, you can click here to edit this entry!
Video Production Service