Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2071 2022-05-21 12:54:37 |
2 update references and layout + 6 word(s) 2077 2022-05-23 05:15:37 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Igelmann, S.; Ferbeyre, G.; Neubauer, H. STAT3 and STAT5 Activation in Solid Cancers. Encyclopedia. Available online: https://encyclopedia.pub/entry/23203 (accessed on 03 July 2024).
Igelmann S, Ferbeyre G, Neubauer H. STAT3 and STAT5 Activation in Solid Cancers. Encyclopedia. Available at: https://encyclopedia.pub/entry/23203. Accessed July 03, 2024.
Igelmann, Sebastian, Gerardo Ferbeyre, Heidi Neubauer. "STAT3 and STAT5 Activation in Solid Cancers" Encyclopedia, https://encyclopedia.pub/entry/23203 (accessed July 03, 2024).
Igelmann, S., Ferbeyre, G., & Neubauer, H. (2022, May 21). STAT3 and STAT5 Activation in Solid Cancers. In Encyclopedia. https://encyclopedia.pub/entry/23203
Igelmann, Sebastian, et al. "STAT3 and STAT5 Activation in Solid Cancers." Encyclopedia. Web. 21 May, 2022.
STAT3 and STAT5 Activation in Solid Cancers
Edit

The Signal Transducer and Activator of Transcription (STAT)3 and 5 proteins are activated by many cytokine receptors to regulate specific gene expression and mitochondrial functions. Their role in cancer is largely context-dependent as they can both act as oncogenes and tumor suppressors.  Activation of Signal Transducer and Activator of Transcription (STAT) proteins has been linked to many human cancers. STATs were initially discovered as latent cytosolic transcription factors that are phosphorylated by the Janus Kinase (JAK) family upon stimulation of membrane-associated cytokine and growth factor receptors. Phosphorylation triggers STAT dimerization and translocation to the nucleus to bind specific promoters and regulate transcription

solid cancers cell cycle apoptosis mitochondria

1. STAT3 and STAT5 in Solid Cancers

The discovery of cancer genes has been propelled by genetic analyses and more recently by next generation DNA sequencing technologies. Combined, these have been identified 127 significantly mutated cancer genes that cover diverse signaling pathways [1]. Mutations acting as drivers in cancer are positively selected during tumor growth and constitute solid proof of the involvement of a particular gene as a driver in the disease. Mutations in STAT3 and STAT5 have been reported in patients with solid cancers, but unlike hyperactivation of the JAK/STAT pathway, STAT3/5 mutations in cancer are relatively infrequent and occur mostly in hematological malignancies.
An overview of reported STAT3/5 mutations in solid cancers, based on data collected from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. Mutations in STAT3 are more prevalent than mutations in STAT5A or STAT5B genes. Noticeably, gastrointestinal cancers have the highest rates of STAT3/5 mutations compared with other solid cancers. Missense mutations tend to cluster within the SH2 domain, where gain-of-function mutations were previously characterized [2][3], as well as within the DNA binding domain and to an extent the N-terminal domain. Interestingly, the STAT3 Tyrosine 640 into Phenylalanine (Y640F) hotspot gain-of-function mutation reported in various lymphoid malignancies has also been detected in patients with liver cancer. Nonsense and frameshift mutations are less frequent and more disperse, likely representing loss-of-function events. Notably, a hotspot frameshift mutation at position Q368 within the DNA binding domain of STAT5B has been reported in 24 patients with various types of carcinoma; this frameshift generates a stop codon shortly after the mutation and is therefore likely to be loss-of-function, although characterization of this mutation has not been performed.
As opposed to mutation rates, STAT3/5 activation is very frequent in human cancers, perhaps reflecting increased cytokine signaling or mutations in cytokine receptors or negative regulators. STAT3/5 activation can be detected using antibodies that measure total levels or activation marks in STAT3/5 proteins (e.g. tyrosine phosphorylation). A better assessment of STAT3/5 activation can be obtained by measuring downstream signaling targets (i.e., mRNA levels of STAT3 [4] and STAT5 [5] target genes). A recent metanalysis of 63 different ones were concluded that STAT3 protein overexpression was significantly associated with a worse 3-year overall survival (OS) (OR = 2.06, 95% CI = 1.57 to 2.71, p < 0.00001) and 5-year OS (OR = 2.00, 95% CI = 1.53 to 2.63, p < 0.00001) in patients with solid tumors [6]. Elevated STAT3 expression was associated with poor prognosis in gastric cancer, lung cancer, gliomas, hepatic cancer, osteosarcoma, prostate cancer and pancreatic cancer. However, high STAT3 protein expression levels predicted a better prognosis for breast cancer [6]. It was mixed data of both STAT3 and phospho-STAT3 (p-STAT3) expression limiting its ability to associate pathway activation to prognosis. It was summarized the data linking activation of STAT3/5 to overall survival in several major human solid cancers identifying the biomarkers used. Taken together, it was clearly showed that STAT3 and STAT5 are important cancer genes despite their relatively low mutation frequency.
STAT3 activation is clearly a factor linked to bad prognosis in patients with lung cancer, liver cancer, renal cell carcinoma (RCC) and gliomas. In other tumors, the association is not significant. In solid tumors, STAT3 activation is more frequent than STAT5 activation although no explanation for this difference was proposed. In prostate cancer, both STAT3 and STAT5 have been associated with castration-resistant disease and proposed as therapeutic targets [7][8]. In colon cancer, the association between p-STAT3 and survival varies. But a high p-STAT3/p-STAT5 ratio indicates bad prognosis [9]. Also in breast cancer, p-STAT5 levels are clearly associated with better prognosis [10]. In liver cancer, STAT5 has ambivalent functions that were recently reviewed by Moriggl and colleagues [11]. Understanding mechanistically how STAT3/5 promote transformation and tumor suppression is important for the eventual design of new treatments. Also, survival data is highly influenced by the response of patients to their treatment and may not always reflect all mechanistic links between STAT3/5 activity and tumor biology. Of note, the effect of any gene is conditioned by the genetic context of gene action. Some genes can clearly exert a tumor suppressor effect in the initial stages of carcinogenesis that is lost when cancer mutations or epigenetic changes inactivate key effectors of these tumor suppressor pathways [12]. Human are usually limited to late stage tumors because it is easier to collect samples at that point. In model systems, including primary cells, organoids and mouse models are thus required for a full understanding of how cancer genes work specifically at early stages in tumorigenesis.

2. Mechanisms of Transformation by STAT3/5 Proteins in Solid Cancers

STAT3 and STAT5 promote tumor progression by regulating the expression of cell cycle, survival and pro-inflammatory genes. In addition, they control mitochondrial functions, metabolism and stemness, as discussed below .

2.1. Cell Cycle and Apoptosis

As transcription factors, STAT3 and STAT5 regulate many genes required for cell cycle progression and cell survival. A major target of the transcriptional control of the mammalian cell cycle is cyclin D. STAT3 regulates cyclin D expression in a complex with CD44 and the acetyltransferase p300. The latter acetylates STAT3 promoting its dimerization, nuclear translocation and binding to the cyclin D promoter [13]. Other cell cycle and survival genes regulated by STAT3 include c-MYC (myc proto-oncogene), B-cell lymphoma 2 (BCL2), BCL2L1/BCL-XL (B-cell lymphoma-extra large), MCL1 (Myeloid Cell Leukemia Sequence 1) and BIRC5/survivin [14]. It was combined ChIPSeq with whole transcriptome profiling in ABC DLBCL (activated B cell-like diffuse large B cell lymphoma) cell lines and revealed that STAT3 activates genes in the Phosphoinositide 3-Kinase (PI3K)/AKT/Mammalian Target of Rapamycin (mTOR) pathway, the Nuclear Factor Kappa-Light-Chain Enhancer of Activated B-Cells (NF-κB) pathway and the cell cycle regulation pathway, while repressing type I interferon signaling genes [15]. STAT5 also regulates the expression of cell cycle and cell survival genes [12] including AKT1 [16], which encodes a pro-survival kinase.

2.2. Inflammation and Innate Immunity

Although the induction of cell proliferation and cell survival genes by STAT3/5 proteins contribute to their pro-cancer activity, in basal-like breast cancers the major genes associated with STAT3 activation control inflammation and the immune response [17]. Of note, inflammation is initially an adaptive response to pathological insults such as oncogenic stimuli, and it therefore exerts a tumor suppressive function. However, dysregulated inflammation in the long term provides a substrate for tumorigenesis [18]. STAT3 alone or in cooperation with NF-κB regulates the expression of many pro-inflammatory genes [19][20][21]. Starved tumor cells activate NF-κB and STAT3 via endoplasmic reticulum (ER) stress and secrete cytokines that stimulate tumor survival and clonogenic capacity [22]. The coactivation of these two transcription factors amplifies pro-inflammatory gene expression driving cancer-associated inflammation [23]. Of interest, the STAT3-NF-κB complex can repress the expression of DNA Damage Inducible Transcript 3 (DDIT3), an inhibitor of CCAAT Enhancer Binding Protein Beta (CEBPβ), another pro-inflammatory transcription factor [24].

Pharmacological agents that limit inflammation have been proposed for cancer prevention [25]. The use of metformin, a drug widely used to control diabetes, has been associated with a dramatic reduction in cancer incidence in many tissues [26]. Although the primary site of action of this drug is in mitochondria, a consequence of its effects is a potent reduction in the activation of NF-κB and STAT3, suggesting that the promising anticancer actions of metformin are related to its ability to curtail pro-inflammatory gene expression [27][28]. In contrast to STAT3, STAT5B inhibits NF-κB activity in the kidney fibroblast cell line COS by competing with coactivators of transcription [29], while it stimulates NF-κB in leukemia cells [30]. These results suggest the involvement of different regulatory mechanisms of STAT5 in hematopoietic cancers compared with solid cancers.

2.3. Mitochondria

In addition to their canonical roles in inflammation and immunity, STAT3 and STAT5 have been shown to localize to mitochondria. The mitochondrial localization of STAT3 is required for its ability to support malignant transformation in murine embryonic fibroblasts and breast cancer cells [31][32][33][34], and mito-STAT3 regulates mitochondrial metabolism and mitochondrial gene expression [33][35][36][37][38][39]. Several have been suggested that STAT3 can be imported to mitochondria after phosphorylation on S727 [32][33] or upon acetylation [40][41]. Other have revealed that STAT3 mitochondrial translocation is mediated by interactions with Heat Shock Protein 22 (HSP22), Gene Associated with Retinoic and Interferon-Induced Mortality 19 (GRIM-19) or Translocase of Outer Mitochondrial Membrane 20 (TOM20) [42][43][44]. The mRNAs coding for some mitochondrial proteins are translated close to or in physical interaction with the import complex TOM [45][46]. The structural motifs mediating those interactions are located in the 3′ and 5′ UTRs of the mRNAs [47][48] and it will be interesting to investigate whether the mRNA of STAT3 also possesses RNA localization signals (zip codes) to localize in close proximity to mitochondria.
Whereas the role of mitochondrial STAT3 has been extensively learned, the role of STAT5 in mitochondria is less clear. The import of STAT5 to mitochondria is regulated by cytokines [31]. Once imported into the mitochondria, STAT5 binds the D-loop of mitochondrial DNA, although no increase in transcription of mitochondrial genes was observed [49]. Mito-STAT5 is also able to interact with the Pyruvate Dehydrogenase Complex (PDC) and was shown to regulate metabolism towards glycolysis, as observed in cells treated with cytokines [31][49]. In the same line, STAT3 was also shown to interact with the PDC in mitochondria [41].

2.4. Reprogramming and Stemness

The role of STAT3 in stem cell biology was initially recognized due to the requirement for the cytokine LIF to maintain pluripotency in cultures of mouse embryonic stem (ES) cells. STAT3 activation mediates the induction or repression of several genes in mouse ES cells including the pluripotency factors Oct4, Klf4, Tfcp2l1 and polycomb proteins [50][51][52]. Many pluripotency factors, such as Homeobox Protein NANOG, are short-lived proteins. STAT3 controls protein stability by inducing the expression of the deubiquitinase Ubiquitin Specific Peptidase 21 (USP21), stabilizing NANOG in mouse ES cells. Induction of ES cell differentiation promotes the Extracellular Signal-Regulated Kinase (ERK)-dependent phosphorylation of USP21 and its dissociation from NANOG, leading to NANOG degradation [53]. STAT3 also plays a role in the reprogramming of somatic cells into induced pluripotent stem (IPS) cells [54] and it has been suggested that its effects depend on the demethylation of pluripotency factor promoters [55]. STAT3 also activates mitochondrial DNA transcription, promoting oxidative phosphorylation during maintenance and induction of pluripotency [56]. It is thus likely that the ability of STAT3 to stimulate stemness also plays a role in its oncogenic activity.
In many tumors, a subpopulation of cells possess a higher malignant capacity. These so-called tumor-initiating cells are suspected to regenerate the tumor after cancer chemotherapy and express many genes commonly expressed in ES cells [57]. It has been shown that STAT3 is required for the formation of tumorspheres and the viability of the cancer stem cell pool in many different tumors [27][28][58][59][60][61][62][63][64][65][66][67][68][69][70][71]. At least in breast cancer, a critical mechanism stimulated by STAT3 to regulate stemness involves genes in fatty acid oxidation [66][67] and the ability of STAT3 to adjust the levels of reactive oxygen species (ROS) produced in mitochondria [67]. In colorectal cancer cells, STAT3 forms a complex with the stem cell marker CD44 and the p300 acetyltransferase. Acetylation of STAT3 by this complex allows dimerization, nuclear translocation and binding to the promoters of genes required for stemness such as c-MYC and TWIST1 [72].
The role of STAT5 in promoting cancer stemness does not affect many cell types and is mostly confined to hematopoietic cancers [73]. However, Nevalainen and colleagues reported that STAT5B induces stem cell properties in prostate cancer cells [74] in line with the increase in nuclear STAT5A/B observed in these tumors in correlation with bad prognosis [8]. Furthermore, transgenic mice with increased expression of prolactin in prostate epithelial cells displayed increases in the basal/stem cell compartment in association with activation of STAT5. This enrichment of stem cells was partially reversed by depletion of Stat5a/b [75]. The pro-stem cell oncogenic effect of STAT5 in the prostate contrasts with its effects in the mammary gland where STAT5 induces cell differentiation [76]. The ETS transcription factor Elf5 (E74-like factor 5) is a target of the prolactin-STAT5 axis and promotes mammary cell differentiation [77][78][79], supporting the tumor suppressive role of STAT5 in the mammary gland.

References

  1. Kandoth, C.; McLellan, M.D.; Vandin, F.; Ye, K.; Niu, B.; Lu, C.; Xie, M.; Zhang, Q.; McMichael, J.F.; Wyczalkowski, M.A.; et al. Mutational landscape and significance across 12 major cancer types. Nature 2013, 502, 333–339.
  2. de Araujo, E.D.; Erdogan, F.; Neubauer, H.A.; Meneksedag-Erol, D.; Manaswiyoungkul, P.; Eram, M.S.; Seo, H.S.; Qadree, A.K.; Israelian, J.; Orlova, A.; et al. Structural and functional consequences of the STAT5B(N642H) driver mutation. Nat. Commun. 2019, 10, 2517.
  3. Koskela, H.L.; Eldfors, S.; Ellonen, P.; van Adrichem, A.J.; Kuusanmaki, H.; Andersson, E.I.; Lagstrom, S.; Clemente, M.J.; Olson, T.; Jalkanen, S.E.; et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N. Engl. J. Med. 2012, 366, 1905–1913.
  4. Carpenter, R.L.; Lo, H.W. STAT3 Target Genes Relevant to Human Cancers. Cancers (Basel) 2014, 6, 897–925.
  5. Basham, B.; Sathe, M.; Grein, J.; McClanahan, T.; D’Andrea, A.; Lees, E.; Rascle, A. In vivo identification of novel STAT5 target genes. Nucleic Acids Res. 2008, 36, 3802–3818.
  6. Wu, P.; Wu, D.; Zhao, L.; Huang, L.; Shen, G.; Huang, J.; Chai, Y. Prognostic role of STAT3 in solid tumors: A systematic review and meta-analysis. Oncotarget 2016, 7, 19863–19883.
  7. Mohanty, S.K.; Yagiz, K.; Pradhan, D.; Luthringer, D.J.; Amin, M.B.; Alkan, S.; Cinar, B. STAT3 and STAT5A are potential therapeutic targets in castration-resistant prostate cancer. Oncotarget 2017, 8, 85997–86010.
  8. Mirtti, T.; Leiby, B.E.; Abdulghani, J.; Aaltonen, E.; Pavela, M.; Mamtani, A.; Alanen, K.; Egevad, L.; Granfors, T.; Josefsson, A.; et al. Nuclear Stat5a/b predicts early recurrence and prostate cancer-specific death in patients treated by radical prostatectomy. Hum. Pathol. 2013, 44, 310–319.
  9. Klupp, F.; Diers, J.; Kahlert, C.; Neumann, L.; Halama, N.; Franz, C.; Schmidt, T.; Lasitschka, F.; Warth, A.; Weitz, J.; et al. Expressional STAT3/STAT5 Ratio is an Independent Prognostic Marker in Colon Carcinoma. Ann. Surg. Oncol. 2015, 22, S1548–S1555.
  10. Peck, A.R.; Witkiewicz, A.K.; Liu, C.; Stringer, G.A.; Klimowicz, A.C.; Pequignot, E.; Freydin, B.; Tran, T.H.; Yang, N.; Rosenberg, A.L.; et al. Loss of nuclear localized and tyrosine phosphorylated Stat5 in breast cancer predicts poor clinical outcome and increased risk of antiestrogen therapy failure. J. Clin. Oncol. 2011, 29, 2448–2458.
  11. Kaltenecker, D.; Themanns, M.; Mueller, K.M.; Spirk, K.; Suske, T.; Merkel, O.; Kenner, L.; Luis, A.; Kozlov, A.; Haybaeck, J.; et al. Hepatic growth hormone-JAK2-STAT5 signalling: Metabolic function, non-alcoholic fatty liver disease and hepatocellular carcinoma progression. Cytokine 2018.
  12. Ferbeyre, G.; Moriggl, R. The role of Stat5 transcription factors as tumor suppressors or oncogenes. Biochim. Biophys. Acta 2011, 1815, 104–114.
  13. Lee, J.L.; Wang, M.J.; Chen, J.Y. Acetylation and activation of STAT3 mediated by nuclear translocation of CD44. J. Cell Biol. 2009, 185, 949–957.
  14. Banerjee, K.; Resat, H. Constitutive activation of STAT3 in breast cancer cells: A review. Int. J. Cancer 2016, 138, 2570–2578.
  15. Lu, L.; Zhu, F.; Zhang, M.; Li, Y.; Drennan, A.C.; Kimpara, S.; Rumball, I.; Selzer, C.; Cameron, H.; Kellicut, A.; et al. Gene regulation and suppression of type I interferon signaling by STAT3 in diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA 2018, 115, E498–E505.
  16. Creamer, B.A.; Sakamoto, K.; Schmidt, J.W.; Triplett, A.A.; Moriggl, R.; Wagner, K.U. Stat5 promotes survival of mammary epithelial cells through transcriptional activation of a distinct promoter in Akt1. Mol. Cell. Biol. 2010, 30, 2957–2970.
  17. Tell, R.W.; Horvath, C.M. Bioinformatic analysis reveals a pattern of STAT3-associated gene expression specific to basal-like breast cancers in human tumors. Proc. Natl. Acad. Sci. USA 2014, 111, 12787–12792.
  18. Karki, R.; Kanneganti, T.D. Diverging inflammasome signals in tumorigenesis and potential targeting. Nat. Rev. Cancer 2019, 19, 197–214.
  19. Martincuks, A.; Andryka, K.; Kuster, A.; Schmitz-Van de Leur, H.; Komorowski, M.; Muller-Newen, G. Nuclear translocation of STAT3 and NF-kappaB are independent of each other but NF-kappaB supports expression and activation of STAT3. Cell. Signal. 2017, 32, 36–47.
  20. Goldstein, I.; Paakinaho, V.; Baek, S.; Sung, M.H.; Hager, G.L. Synergistic gene expression during the acute phase response is characterized by transcription factor assisted loading. Nat. Commun. 2017, 8, 1849.
  21. Grivennikov, S.I.; Karin, M. Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010, 21, 11–19.
  22. Yoon, S.; Woo, S.U.; Kang, J.H.; Kim, K.; Shin, H.J.; Gwak, H.S.; Park, S.; Chwae, Y.J. NF-kappaB and STAT3 cooperatively induce IL6 in starved cancer cells. Oncogene 2012, 31, 3467–3481.
  23. Atsumi, T.; Singh, R.; Sabharwal, L.; Bando, H.; Meng, J.; Arima, Y.; Yamada, M.; Harada, M.; Jiang, J.J.; Kamimura, D.; et al. Inflammation amplifier, a new paradigm in cancer biology. Cancer Res. 2014, 74, 8–14.
  24. Canino, C.; Luo, Y.; Marcato, P.; Blandino, G.; Pass, H.I.; Cioce, M. A STAT3-NFkB/DDIT3/CEBPbeta axis modulates ALDH1A3 expression in chemoresistant cell subpopulations. Oncotarget 2015, 6, 12637–12653.
  25. Thi, H.T.H.; Hong, S. Inflammasome as a Therapeutic Target for Cancer Prevention and Treatment. J. Cancer Prev. 2017, 22, 62–73.
  26. Pollak, M.N. Investigating metformin for cancer prevention and treatment: The end of the beginning. Cancer Discov. 2012, 2, 778–790.
  27. Hirsch, H.A.; Iliopoulos, D.; Struhl, K. Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc. Natl. Acad. Sci. USA 2013, 110, 972–977.
  28. Deschenes-Simard, X.; Parisotto, M.; Rowell, M.C.; Le Calve, B.; Igelmann, S.; Moineau-Vallee, K.; Saint-Germain, E.; Kalegari, P.; Bourdeau, V.; Kottakis, F.; et al. Circumventing senescence is associated with stem cell properties and metformin sensitivity. Aging Cell 2019.
  29. Luo, G.; Yu-Lee, L. Stat5b inhibits NFkappaB-mediated signaling. Mol. Endocrinol. 2000, 14, 114–123.
  30. Kawashima, T.; Murata, K.; Akira, S.; Tonozuka, Y.; Minoshima, Y.; Feng, S.; Kumagai, H.; Tsuruga, H.; Ikeda, Y.; Asano, S.; et al. STAT5 induces macrophage differentiation of M1 leukemia cells through activation of IL-6 production mediated by NF-kappaB p65. J. Immunol. 2001, 167, 3652–3660.
  31. Chueh, F.Y.; Leong, K.F.; Yu, C.L. Mitochondrial translocation of signal transducer and activator of transcription 5 (STAT5) in leukemic T cells and cytokine-stimulated cells. Biochem. Biophys. Res. Commun. 2010, 402, 778–783.
  32. Gough, D.J.; Corlett, A.; Schlessinger, K.; Wegrzyn, J.; Larner, A.C.; Levy, D.E. Mitochondrial STAT3 supports Ras-dependent oncogenic transformation. Science 2009, 324, 1713–1716.
  33. Wegrzyn, J.; Potla, R.; Chwae, Y.J.; Sepuri, N.B.; Zhang, Q.; Koeck, T.; Derecka, M.; Szczepanek, K.; Szelag, M.; Gornicka, A.; et al. Function of mitochondrial Stat3 in cellular respiration. Science 2009, 323, 793–797.
  34. Zhang, Q.; Raje, V.; Yakovlev, V.A.; Yacoub, A.; Szczepanek, K.; Meier, J.; Derecka, M.; Chen, Q.; Hu, Y.; Sisler, J.; et al. Mitochondrial localized Stat3 promotes breast cancer growth via phosphorylation of serine 727. J. Biol. Chem. 2013, 288, 31280–31288.
  35. Macias, E.; Rao, D.; Carbajal, S.; Kiguchi, K.; DiGiovanni, J. Stat3 binds to mtDNA and regulates mitochondrial gene expression in keratinocytes. J. Investig. Dermatol. 2014, 134, 1971–1980.
  36. Rincon, M.; Pereira, F.V. A New Perspective: Mitochondrial Stat3 as a Regulator for Lymphocyte Function. Int. J. Mol. Sci. 2018, 19, 1656.
  37. Avalle, L.; Poli, V. Nucleus, Mitochondrion, or Reticulum? STAT3 a La Carte. Int. J. Mol. Sci. 2018, 19, 2820.
  38. Luo, D.; Fraga-Lauhirat, M.; Millings, J.; Ho, C.; Villarreal, E.M.; Fletchinger, T.C.; Bonfiglio, J.V.; Mata, L.; Nemesure, M.D.; Bartels, L.E.; et al. Phospho-valproic acid (MDC-1112) suppresses glioblastoma growth in preclinical models through the inhibition of STAT3 phosphorylation. Carcinogenesis 2019.
  39. Sala, D.; Cunningham, T.J.; Stec, M.J.; Etxaniz, U.; Nicoletti, C.; Dall’Agnese, A.; Puri, P.L.; Duester, G.; Latella, L.; Sacco, A. The Stat3-Fam3a axis promotes muscle stem cell myogenic lineage progression by inducing mitochondrial respiration. Nat. Commun. 2019, 10, 1796.
  40. Bernier, M.; Paul, R.K.; Martin-Montalvo, A.; Scheibye-Knudsen, M.; Song, S.; He, H.J.; Armour, S.M.; Hubbard, B.P.; Bohr, V.A.; Wang, L.; et al. Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 protein. J. Biol. Chem. 2011, 286, 19270–19279.
  41. Xu, Y.S.; Liang, J.J.; Wang, Y.; Zhao, X.J.; Xu, L.; Xu, Y.Y.; Zou, Q.C.; Zhang, J.M.; Tu, C.E.; Cui, Y.G.; et al. STAT3 Undergoes Acetylation-dependent Mitochondrial Translocation to Regulate Pyruvate Metabolism. Sci. Rep. 2016, 6, 39517.
  42. Boengler, K.; Hilfiker-Kleiner, D.; Heusch, G.; Schulz, R. Inhibition of permeability transition pore opening by mitochondrial STAT3 and its role in myocardial ischemia/reperfusion. Basic Res. Cardiol. 2010, 105, 771–785.
  43. Tammineni, P.; Anugula, C.; Mohammed, F.; Anjaneyulu, M.; Larner, A.C.; Sepuri, N.B. The import of the transcription factor STAT3 into mitochondria depends on GRIM-19, a component of the electron transport chain. J. Biol. Chem. 2013, 288, 4723–4732.
  44. Qiu, H.; Lizano, P.; Laure, L.; Sui, X.; Rashed, E.; Park, J.Y.; Hong, C.; Gao, S.; Holle, E.; Morin, D.; et al. H11 kinase/heat shock protein 22 deletion impairs both nuclear and mitochondrial functions of STAT3 and accelerates the transition into heart failure on cardiac overload. Circulation 2011, 124, 406–415.
  45. Gadir, N.; Haim-Vilmovsky, L.; Kraut-Cohen, J.; Gerst, J.E. Localization of mRNAs coding for mitochondrial proteins in the yeast Saccharomyces cerevisiae. RNA 2011, 17, 1551–1565.
  46. Fox, T.D. Mitochondrial protein synthesis, import, and assembly. Genetics 2012, 192, 1203–1234.
  47. Zhang, Y.; Chen, Y.; Gucek, M.; Xu, H. The mitochondrial outer membrane protein MDI promotes local protein synthesis and mtDNA replication. EMBO J. 2016, 35, 1045–1057.
  48. Hansen, K.G.; Herrmann, J.M. Transport of Proteins into Mitochondria. Protein J. 2019.
  49. Richard, A.J.; Hang, H.; Stephens, J.M. Pyruvate dehydrogenase complex (PDC) subunits moonlight as interaction partners of phosphorylated STAT5 in adipocytes and adipose tissue. J. Biol. Chem. 2017, 292, 19733–19742.
  50. Kidder, B.L.; Yang, J.; Palmer, S. Stat3 and c-Myc genome-wide promoter occupancy in embryonic stem cells. PLoS ONE 2008, 3, e3932.
  51. Hall, J.; Guo, G.; Wray, J.; Eyres, I.; Nichols, J.; Grotewold, L.; Morfopoulou, S.; Humphreys, P.; Mansfield, W.; Walker, R.; et al. Oct4 and LIF/Stat3 additively induce Kruppel factors to sustain embryonic stem cell self-renewal. Cell Stem Cell 2009, 5, 597–609.
  52. Ye, S.; Li, P.; Tong, C.; Ying, Q.L. Embryonic stem cell self-renewal pathways converge on the transcription factor Tfcp2l1. EMBO J. 2013, 32, 2548–2560.
  53. Jin, J.; Liu, J.; Chen, C.; Liu, Z.; Jiang, C.; Chu, H.; Pan, W.; Wang, X.; Zhang, L.; Li, B.; et al. The deubiquitinase USP21 maintains the stemness of mouse embryonic stem cells via stabilization of Nanog. Nat. Commun. 2016, 7, 13594.
  54. Yang, J.; van Oosten, A.L.; Theunissen, T.W.; Guo, G.; Silva, J.C.; Smith, A. Stat3 activation is limiting for reprogramming to ground state pluripotency. Cell Stem Cell 2010, 7, 319–328.
  55. Tang, Y.; Luo, Y.; Jiang, Z.; Ma, Y.; Lin, C.J.; Kim, C.; Carter, M.G.; Amano, T.; Park, J.; Kish, S.; et al. Jak/Stat3 signaling promotes somatic cell reprogramming by epigenetic regulation. Stem Cells 2012, 30, 2645–2656.
  56. Carbognin, E.; Betto, R.M.; Soriano, M.E.; Smith, A.G.; Martello, G. Stat3 promotes mitochondrial transcription and oxidative respiration during maintenance and induction of naive pluripotency. EMBO J. 2016, 35, 618–634.
  57. Ben-Porath, I.; Thomson, M.W.; Carey, V.J.; Ge, R.; Bell, G.W.; Regev, A.; Weinberg, R.A. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 2008, 40, 499–507.
  58. Zhou, J.; Wulfkuhle, J.; Zhang, H.; Gu, P.; Yang, Y.; Deng, J.; Margolick, J.B.; Liotta, L.A.; Petricoin, E., 3rd; Zhang, Y. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc. Natl. Acad. Sci. USA 2007, 104, 16158–16163.
  59. Marotta, L.L.; Almendro, V.; Marusyk, A.; Shipitsin, M.; Schemme, J.; Walker, S.R.; Bloushtain-Qimron, N.; Kim, J.J.; Choudhury, S.A.; Maruyama, R.; et al. The JAK2/STAT3 signaling pathway is required for growth of CD44(+)CD24(-) stem cell-like breast cancer cells in human tumors. J. Clin. Investig. 2011, 121, 2723–2735.
  60. Iliopoulos, D.; Hirsch, H.A.; Struhl, K. An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell 2009, 139, 693–706.
  61. Gong, A.H.; Wei, P.; Zhang, S.; Yao, J.; Yuan, Y.; Zhou, A.D.; Lang, F.F.; Heimberger, A.B.; Rao, G.; Huang, S. FoxM1 Drives a Feed-Forward STAT3-Activation Signaling Loop That Promotes the Self-Renewal and Tumorigenicity of Glioblastoma Stem-like Cells. Cancer Res. 2015, 75, 2337–2348.
  62. Peng, L.; Jiang, D. Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS ONE 2018, 13, e0205918.
  63. Man, J.; Yu, X.; Huang, H.; Zhou, W.; Xiang, C.; Huang, H.; Miele, L.; Liu, Z.; Bebek, G.; Bao, S.; et al. Hypoxic Induction of Vasorin Regulates Notch1 Turnover to Maintain Glioma Stem-like Cells. Cell Stem Cell 2018, 22, 104–118.e6.
  64. Jiao, J.; Zhang, R.; Li, Z.; Yin, Y.; Fang, X.; Ding, X.; Cai, Y.; Yang, S.; Mu, H.; Zong, D.; et al. Nuclear Smad6 promotes gliomagenesis by negatively regulating PIAS3-mediated STAT3 inhibition. Nat. Commun. 2018, 9, 2504.
  65. He, W.; Wu, J.; Shi, J.; Huo, Y.M.; Dai, W.; Geng, J.; Lu, P.; Yang, M.W.; Fang, Y.; Wang, W.; et al. IL22RA1/STAT3 Signaling Promotes Stemness and Tumorigenicity in Pancreatic Cancer. Cancer Res. 2018, 78, 3293–3305.
  66. Wang, T.; Fahrmann, J.F.; Lee, H.; Li, Y.J.; Tripathi, S.C.; Yue, C.; Zhang, C.; Lifshitz, V.; Song, J.; Yuan, Y.; et al. JAK/STAT3-Regulated Fatty Acid beta-Oxidation Is Critical for Breast Cancer Stem Cell Self-Renewal and Chemoresistance. Cell Metab. 2018, 27, 136–150.
  67. Kitajima, S.; Yoshida, A.; Kohno, S.; Li, F.; Suzuki, S.; Nagatani, N.; Nishimoto, Y.; Sasaki, N.; Muranaka, H.; Wan, Y.; et al. The RB-IL-6 axis controls self-renewal and endocrine therapy resistance by fine-tuning mitochondrial activity. Oncogene 2017, 36, 5145–5157.
  68. Chen, M.W.; Yang, S.T.; Chien, M.H.; Hua, K.T.; Wu, C.J.; Hsiao, S.M.; Lin, H.; Hsiao, M.; Su, J.L.; Wei, L.H. The STAT3-miRNA-92-Wnt Signaling Pathway Regulates Spheroid Formation and Malignant Progression in Ovarian Cancer. Cancer Res. 2017, 77, 1955–1967.
  69. Schroeder, A.; Herrmann, A.; Cherryholmes, G.; Kowolik, C.; Buettner, R.; Pal, S.; Yu, H.; Muller-Newen, G.; Jove, R. Loss of androgen receptor expression promotes a stem-like cell phenotype in prostate cancer through STAT3 signaling. Cancer Res. 2014, 74, 1227–1237.
  70. Tang, Y.; Kitisin, K.; Jogunoori, W.; Li, C.; Deng, C.X.; Mueller, S.C.; Ressom, H.W.; Rashid, A.; He, A.R.; Mendelson, J.S.; et al. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling. Proc. Natl. Acad. Sci. USA 2008, 105, 2445–2450.
  71. Thiagarajan, P.S.; Zheng, Q.; Bhagrath, M.; Mulkearns-Hubert, E.E.; Myers, M.G.; Lathia, J.D.; Reizes, O. STAT3 activation by leptin receptor is essential for TNBC stem cell maintenance. Endocr. Relat. Cancer 2017, 24, 415–426.
  72. Su, Y.J.; Lai, H.M.; Chang, Y.W.; Chen, G.Y.; Lee, J.L. Direct reprogramming of stem cell properties in colon cancer cells by CD44. EMBO J. 2011, 30, 3186–3199.
  73. Schepers, H.; van Gosliga, D.; Wierenga, A.T.; Eggen, B.J.; Schuringa, J.J.; Vellenga, E. STAT5 is required for long-term maintenance of normal and leukemic human stem/progenitor cells. Blood 2007, 110, 2880–2888.
  74. Talati, P.G.; Gu, L.; Ellsworth, E.M.; Girondo, M.A.; Trerotola, M.; Hoang, D.T.; Leiby, B.; Dagvadorj, A.; McCue, P.A.; Lallas, C.D.; et al. Jak2-Stat5a/b Signaling Induces Epithelial-to-Mesenchymal Transition and Stem-Like Cell Properties in Prostate Cancer. Am. J. Pathol. 2015, 185, 2505–2522.
  75. Boutillon, F.; Pigat, N.; Sala, L.S.; Reyes-Gomez, E.; Moriggl, R.; Guidotti, J.E.; Goffin, V. STAT5a/b Deficiency Delays, but does not Prevent, Prolactin-Driven Prostate Tumorigenesis in Mice. Cancers (Basel) 2019, 11, 929.
  76. Liu, X.; Robinson, G.W.; Wagner, K.U.; Garrett, L.; Wynshaw-Boris, A.; Hennighausen, L. Stat5a is mandatory for adult mammary gland development and lactogenesis. Genes Dev. 1997, 11, 179–186.
  77. Oakes, S.R.; Naylor, M.J.; Asselin-Labat, M.L.; Blazek, K.D.; Gardiner-Garden, M.; Hilton, H.N.; Kazlauskas, M.; Pritchard, M.A.; Chodosh, L.A.; Pfeffer, P.L.; et al. The ETS transcription factor Elf5 specifies mammary alveolar cell fate. Genes Dev. 2008, 22, 581–586.
  78. Metser, G.; Shin, H.Y.; Wang, C.; Yoo, K.H.; Oh, S.; Villarino, A.V.; O’Shea, J.J.; Kang, K.; Hennighausen, L. An autoregulatory enhancer controls mammary-specific STAT5 functions. Nucleic Acids Res. 2016, 44, 1052–1063.
  79. Stute, P.; Sielker, S.; Wood, C.E.; Register, T.C.; Lees, C.J.; Dewi, F.N.; Williams, J.K.; Wagner, J.D.; Stefenelli, U.; Cline, J.M. Life stage differences in mammary gland gene expression profile in non-human primates. Breast Cancer Res. Treat. 2012, 133, 617–634.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 280
Revisions: 2 times (View History)
Update Date: 23 May 2022
1000/1000
Video Production Service