Functional Effects of TGF-β3: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

Transforming growth factor-beta 3 (TGF-β3) is a ubiquitously expressed multifunctional cytokine involved in a range of physiological and pathological conditions, including embryogenesis, cell cycle regulation, immunoregulation, and fibrogenesis. 

  • cell cycle regulation
  • chemotherapy
  • fibrosis

1. Immunoregulation and Inflammation

TGF-βs (Transforming growth factor-beta) are expressed by immune cells and are thought to play an important immunoregulatory role. TGF-β1 is the most extensively described isoform in regard to immunity, mainly because TGF-β1 null mice develop a fatal systemic autoimmune disease [1]. While the role of TGF-β2 is currently considered to be insignificant in the regulation of the immune system because of its negligible expression in immune cells, TGF-β3 is emerging as a potentially important immunoregulator with functions that differ from those of TGF-β1 in many contexts [2].
Several cells of the immune system have been reported to produce TGF-β3 either at the mRNA or protein level, and the role of TGF-β3 has been reported to be both pro- and anti-inflammatory. Interleukin-17-producing helper T cells (Th17 cells) are generated from naïve CD4+ T cells via several cytokines, including TGF-β. Lee et al. found that while generation with TGF-β1 produced a type of Th17 cell that did not readily induce autoimmune disease, TGF-β3 generated a functionally distinct type of Th17 cell that was highly pathogenic [3]. In B cells, TGF-β3 is thought to enhance antibody production under certain circumstances and can possess bifunctional effects depending on concentration [4].
The anti-inflammatory role of TGF-β3 has been highlighted through its ability to inhibit differentiation of forkhead box P3-expressing CD4+ T cells [5], its potential to inhibit B cell proliferation and antibody production [6], and its role in lymphocyte-activation gene 3+ regulatory T cells (Treg) mediated immune suppression [7]. TGF-β3 was also observed to suppress the development of germinal center B cells and thereby inhibiting the T cell-dependent immune response, a function which was not associated with TGF-β1 [4].

2. Fibrosis and Wound Healing

Radiation fibrosis is a late complication of radiotherapy, with potentially debilitating effects. While it is widely accepted that the action of TGF-β1 is pro-fibrotic, and TGF-β2 is generally believed to aid in this action, conflicting reports exist regarding TGF-β3 [8][9][10][11]. Several studies have reported pro-fibrotic effects of TGF-β3: Sun et al. registered upregulation of TGF-β2 and TGF-β3, but not TGF-β1, in human idiopathic pulmonary fibrosis and non-alcoholic fatty liver disease. In addition, they demonstrated pro-fibrotic effects of TGF-β3 in mouse models of these fibrotic diseases [12]. Guo et al. proposed that TGF-β3 promoted liver fibrosis by downregulation of MMP13 [13]. Reggio et al. reported an increased expression of extracellular matrix and inflammatory genes after treating cultured adipocytes and endothelial cells with TGF-β3 [14].
Others, however, have argued the regulatory or anti-fibrotic roles of TGF-β3 [15][16][17][18]. Although they detected the increased expression of TGF-β3 in tissue samples from human myocardial infarction patients, Xue et al. found that TGF-β3 suppressed cardiac fibroblasts and decreased fibrotic markers in an in vitro model [19]. Wu et al. concluded that transplanted mesenchymal stem cells reduced skin fibrosis through the action of TGF-β3 [20]. Escansany et al. observed renal fibrosis in TGF-β3 deficient mice, in addition to impaired lipid metabolism [21].
Wound healing and scar formation are closely tied to the fibrotic response. Early studies indicated that scarless healing observed in mammalian embryos was due to elevated levels of TGF-β3 in fetal tissue [22][23]. Furthermore, Shah et al. demonstrated that cutaneous scarring in rats was reduced by the addition of exogenous TGF-β3 or by the inhibition of TGF-β1 or -β2 [24]. Indeed, recombinant human TGF-β3 under the trade name Avotermin was developed as an anti-scarring agent, showing potential to provide an accelerated and permanent improvement in scarring in several phase I/II trials [25][26][27], but failed the phase III trial [28]. Despite the clinical failure of Avotermin, several more recent studies have supported TGF-β3′s anti-scarring effects [20][29][30][31][32].

3. Growth Regulation and Carcinogenesis

Shortly after its discovery, TGF-β1 was identified as a growth stimulator in fibroblasts, but a potent growth inhibitor in other cell types, including epithelial, lymphoid, and endothelial cells [33]. Early studies indicated identical functions for TGF-β2 and TGF-β3, but later research has elucidated isoform-specific variations in the growth-regulatory effects of TGF-βs. The addition of exogenous TGF-β3, but not TGF-β1, increased chondrocyte proliferation in neonatal murine cartilage in vitro [34]. Similarly, TGF-β3 increased chondrocyte proliferation in murine cranial suture-derived mesenchymal cells in vitro, while TGF-β1 had the opposite effect [35]. In contrast, mesenchymal stem cells were found to inhibit keloid fibroblast proliferation in vitro through a TGF-β3-dependent mechanism [20], and the addition of exogenous TGF-β3 reduced the proliferation of cultured taste bud epithelial cells [36]. Treatment with TGF-β3 also inhibited the proliferation of smooth muscle cells in vitro [37][38].
When considering the role of TGF-β3 in radiation cancer therapy, it is also of interest to consider its role in the disease itself. Research regarding the role of TGF-β3 in carcinogenesis and cancer progression is in many cases obscured by an assumption that the three TGF-β isoforms have identical functions and are interchangeable. This assumption is not necessarily true and can often be misleading. Furthermore, the majority of data referenced regarding the tumorigenic role of TGF-β3 originates from protein or mRNA analyses of tumor biopsies and are thus correlative [39]. Considerable caution should be applied before interpreting the data as causative.
In an animal model of cutaneous melanoma, TGF-β3, unlike TGF-β1 and TGF-β2, was induced in cells adjacent to tumor tissue by factors released by the malignant cells, suggesting a responsive rather than a causative role of TGF-β3 [40]. Another study observed decreased TGF-β3 expression in the skin overlying malignant melanoma and suggested that melanoma cells either suppressed TGF-β3 expression or that the lack of TGF-β3 expression itself promoted melanocyte proliferation [41].
In several studies identifying genes associated with prognosis in breast cancer patients, increased TGF-β3 mRNA expression was correlated with a longer interval to distal metastases [42][43][44]. However, increased TGF-β3 protein levels have been observed to correlate with the incidence of lymph node metastases [45], a decrease in survival time [46], and overall survival [47] in breast cancer patients. One study found that the increased expression of TGF-β3 mRNA could lead to ovarian carcinogenesis and tumor progression [48], while another concluded that the increased TGF-β3 expression induced by progestin treatment was associated with a lower risk of developing ovarian cancer in macaques [49]. An analysis of osteosarcoma biopsies revealed a correlation between TGF-β3 protein content and lung metastasis incidence [50]. Leiomyoma samples have been shown to have a higher expression of TGF-β3 mRNA and protein than normal myometrium, and the addition of TGF-β3 increased the proliferation of cultured leiomyoma cells [38][51][52].

This entry is adapted from the peer-reviewed paper 10.3390/ijms24087614

References

  1. Kulkarni, A.B.; Huh, C.G.; Becker, D.; Geiser, A.; Lyght, M.; Flanders, K.C.; Roberts, A.B.; Sporn, M.B.; Ward, J.M.; Karlsson, S. Transforming Growth Factor Β1 Null Mutation in Mice Causes Excessive Inflammatory Response and Early Death. Proc. Natl. Acad. Sci. USA 1993, 90, 770–774.
  2. Komai, T.; Okamura, T.; Inoue, M.; Yamamoto, K.; Fujio, K. Reevaluation of Pluripotent Cytokine TGF-Β3 in Immunity. Int. J. Mol. Sci. 2018, 19, 2261.
  3. Lee, Y.; Awasthi, A.; Yosef, N.; Quintana, F.J.; Xiao, S.; Peters, A.; Wu, C.; Kleinewietfeld, M.; Kunder, S.; Hafler, D.A.; et al. Induction and Molecular Signature of Pathogenic T H 17 Cells. Nat. Immunol. 2012, 13, 991–999.
  4. Komai, T.; Inoue, M.; Okamura, T.; Morita, K.; Iwasaki, Y.; Sumitomo, S.; Shoda, H.; Yamamoto, K.; Fujio, K. Transforming Growth Factor-β and Interleukin-10 Synergistically Regulate Humoral Immunity via Modulating Metabolic Signals. Front. Immunol. 2018, 9, 1364.
  5. Shah, S.; Qiao, L. Resting B Cells Expand a CD4+CD25+Foxp3+ Treg Population via TGF-Β3. Eur. J. Immunol. 2008, 38, 2488–2498.
  6. Tsuchida, Y.; Sumitomo, S.; Ishigaki, K.; Suzuki, A.; Kochi, Y.; Tsuchiya, H.; Ota, M.; Komai, T.; Inoue, M.; Morita, K.; et al. TGF-Β3 Inhibits Antibody Production by Human B Cells. PLoS ONE 2017, 12, e0169646.
  7. Okamura, T.; Sumitomo, S.; Morita, K.; Iwasaki, Y.; Inoue, M.; Nakachi, S.; Komai, T.; Shoda, H.; Miyazaki, J.I.; Fujio, K.; et al. TGF-Β3-Expressing CD4+CD25-LAG3+ Regulatory T Cells Control Humoral Immune Responses. Nat. Commun. 2015, 6, 6329.
  8. Martin, M.; Lefaix, J.-L.; Sylvie, D. TGF-Beta1 and radiation fibrosis: A master switch and a specific therapeutic target? Int. J. Radiat. Oncol. 2000, 47, 277–290.
  9. Durani, P.; Occleston, N.; O’Kane, S.; Ferguson, M.W.J. Avotermin: A Novel Antiscarring Agent. Int. J. Low. Extrem. Wounds 2008, 7, 160–168.
  10. Kim, K.K.; Sheppard, D.; Chapman, H.A. TGF-β1 signaling and tissue fibrosis. Cold Spring Harb. Perspect. Biol. 2018, 10, a022293.
  11. Frangogiannis, N.G. Transforming Growth Factor–ß in Tissue Fibrosis. J. Exp. Med. 2020, 217, e20190103.
  12. Sun, T.; Huang, Z.; Liang, W.C.; Yin, J.; Lin, W.Y.; Wu, J.; Vernes, J.M.; Lutman, J.; Caplazi, P.; Jeet, S.; et al. TGFβ2 and TGFβ3 Isoforms Drive Fibrotic Disease Pathogenesis. Sci. Transl. Med. 2021, 13, eabe0407.
  13. Guo, J.; Liu, W.; Zeng, Z.; Lin, J.; Zhang, X.; Chen, L. Tgfb3 and Mmp13 Regulated the Initiation of Liver Fibrosis Progression as Dynamic Network Biomarkers. J. Cell. Mol. Med. 2021, 25, 867–879.
  14. Reggio, S.; Rouault, C.; Poitou, C.; Bichet, J.-C.; Prifti, E.; Bouillot, J.-L.; Rizkalla, S.; Lacasa, D.; Tordjman, J.; Clément, K. Increased Basement Membrane Components in Adipose Tissue During Obesity: Links With TGFβ and Metabolic Phenotypes. J. Clin. Endocrinol. Metab. 2016, 101, 2578–2587.
  15. Serini, G.; Gabbiani, G. Modulation of alpha-smooth muscle actin expression in fibroblasts by transforming growth factor-beta isoforms: An in vivo and in vitro study. Wound Repair Regen. 1996, 3, 278–287.
  16. Loewen, M.S.; Walner, D.L.; Caldarelli, D.D. Improved Airway Healing Using Transforming Growth Factor Beta-3 In a Rabbit Model. Wound Repair Regen. 2001, 9, 44–49.
  17. Ask, K.; Bonniaud, P.; Maass, K.; Eickelberg, O.; Margetts, P.J.; Warburton, D.; Groffen, J.; Gauldie, J.; Kolb, M. Progressive Pulmonary Fibrosis Is Mediated by TGF-β Isoform 1 but Not TGF-Β3. Int. J. Biochem. Cell Biol. 2008, 40, 484–495.
  18. Wilson, S.E. TGF Beta −1, −2 and −3 in the Modulation of Fibrosis in the Cornea and Other Organs. Exp. Eye Res. 2021, 207, 108594.
  19. Xue, K.; Zhang, J.; Li, C.; Li, J.; Wang, C.; Zhang, Q.; Chen, X.; Yu, X.; Sun, L.; Yu, X. The Role and Mechanism of Transforming Growth Factor Beta 3 in Human Myocardial Infarction-Induced Myocardial Fibrosis. J. Cell. Mol. Med. 2019, 23, 4229–4243.
  20. Wu, Y.; Peng, Y.; Gao, D.; Feng, C.; Yuan, X.; Li, H.; Wang, Y.; Yang, L.; Huang, S.; Fu, X. Mesenchymal stem cells suppress fibroblast proliferation and reduce skin fibrosis through a TGF-β3-dependent activation. Int. J. Low. Extrem. Wounds 2015, 14, 50–62.
  21. Escasany, E.; Lanzón, B.; García-Carrasco, A.; Izquierdo-Lahuerta, A.; Torres, L.; Corrales, P.; Rodríguez, A.E.R.; Luis-Lima, S.; Álvarez, C.M.; Ruperez, F.J.; et al. Transforming Growth Factor β 3 Deficiency Promotes Defective Lipid Metabolism and Fibrosis in Murine Kidney. Dis. Model. Mech. 2021, 14, dmm048249.
  22. Whitby, D.J.; Ferguson, M.W.J. The Extracellular Matrix of Lip Wounds in Fetal, Neonatal and Adult Mice. Development 1991, 112, 651–668.
  23. Chen, W.; Fu, X.; Ge, S.; Sun, T.; Zhou, G.; Jiang, D.; Sheng, Z. Ontogeny of Expression of Transforming Growth Factor-β and Its Receptors and Their Possible Relationship with Scarless Healing in Human Fetal Skin. Wound Repair Regen. 2005, 13, 68–75.
  24. Shah, M.; Foreman, D.M.; Ferguson, M.W. Neutralisation of TGF-beta 1 and TGF-beta 2 or exogenous addition of TGF-beta 3 to cutaneous rat wounds reduces scarring. J. Cell Sci. 1995, 108, 985–1002.
  25. Ferguson, M.W.; Duncan, J.; Bond, J.; Bush, J.; Durani, P.; So, K.; Taylor, L.; Chantrey, J.; Mason, T.; James, G.; et al. Prophylactic Administration of Avotermin for Improvement of Skin Scarring: Three Double-Blind, Placebo-Controlled, Phase I/II Studies. Lancet 2009, 373, 1264–1274.
  26. Bush, J.; Duncan, J.A.L.; Bond, J.S.; Durani, P.; So, K.; Mason, T.; O’Kane, S.; Ferguson, M.W.J. Scar-Improving Efficacy of Avotermin Administered into the Wound Margins of Skin Incisions as Evaluated by a Randomized, Double-Blind, Placebo-Controlled, Phase II Clinical Trial. Plast. Reconstr. Surg. 2010, 126, 1604–1615.
  27. So, K.; McGrouther, D.A.; Bush, J.A.; Durani, P.; Taylor, L.; Skotny, G.; Mason, T.; Metcalfe, A.; Okane, S.; Ferguson, M.W.J. Avotermin for Scar Improvement Following Scar Revision Surgery: A Randomized, Double-Blind, within-Patient, Placebo-Controlled, Phase II Clinical Trial. Plast. Reconstr. Surg. 2011, 128, 163–172.
  28. Juvista EU Phase 3 Trial Results. Available online: https://www.fiercebiotech.com/biotech/juvista-eu-phase-3-trial-results (accessed on 13 March 2023).
  29. Chang, Z.; Kishimoto, Y.; Hasan, A.; Welham, N.V. TGF-Β3 Modulates the Inflammatory Environment and Reduces Scar Formation Following Vocal Fold Mucosal Injury in Rats. Dis. Model. Mech. 2014, 7, 83–91.
  30. Gómez-Gil, V.; Pascual, G.; Pérez-Köhler, B.; Cifuentes, A.; Buján, J.; Bellón, J.M. Involvement of Transforming Growth Factor-Β3 and Betaglycan in the Cytoarchitecture of Postoperative Omental Adhesions. J. Surg. Res. 2014, 187, 699–711.
  31. Karamichos, D.; Hutcheon, A.E.K.; Zieske, J.D. Reversal of Fibrosis by TGF-Β3 in a 3D in Vitro Model. Exp. Eye Res. 2014, 124, 31–36.
  32. Honardoust, D.; Varkey, M.; Marcoux, Y.; Shankowsky, H.A.; Tredget, E.E. Reduced decorin, fibromodulin, and transforming growth factor-β3 in deep dermis leads to hypertrophic scarring. J. Burn. Care Res. 2012, 33, 218–227.
  33. Barnard, J.A.; Lyons, R.M.; Moses, H.L. The Cell Biology of Transforming Growth Factor β. Biochimica et Biophysica Acta (BBA)-Rev. Cancer 1990, 1032, 79–87.
  34. Martinez-Alvernia, E.A.; Rudnick, J.A.; Mankarious, L.A. Transforming growth factor β3 increases chondrocyte proliferation and decreases apoptosis in murine cricoid cartilage in vitro. Otolaryngol. Head Neck Surg. 2008, 138, 435–440.
  35. James, A.W.; Xu, Y.; Lee, J.K.; Wang, R.; Longaker, M.T. Differential Effects of TGF-Β1 and TGF-Β3 on Chondrogenesis in Posterofrontal Cranial Suture-Derived Mesenchymal Cells in vitro. Plast. Reconstr. Surg. 2009, 123, 31–43.
  36. Nakamura, S.I.; Kawai, T.; Kamakura, T.; Ookura, T. TGF-Β3 Is Expressed in Taste Buds and Inhibits Proliferation of Primary Cultured Taste Epithelial Cells. Vitr. Cell. Dev. Biol.-Anim. 2010, 46, 36–44.
  37. Zeng, L.; Li, Y.; Yang, J.; Wang, G.; Margariti, A.; Xiao, Q.; Zampetaki, A.; Yin, X.; Mayr, M.; Mori, K.; et al. XBP 1-Deficiency Abrogates Neointimal Lesion of Injured Vessels via Cross Talk with the PDGF Signaling. Arter. Thromb. Vasc. Biol. 2015, 35, 2134–2144.
  38. Miyashita-Ishiwata, M.; El Sabeh, M.; Reschke, L.D.; Afrin, S.; Borahay, M.A. Hypoxia Induces Proliferation via NOX4-Mediated Oxidative Stress and TGF-Β3 Signaling in Uterine Leiomyoma Cells. Free. Radic. Res. 2022, 56, 163–172.
  39. Laverty, H.G.; Wakefield, L.M.; Occleston, N.L.; O’Kane, S.; Ferguson, M.W.J. TGF-Β3 and Cancer: A Review. Cytokine Growth Factor Rev. 2009, 20, 305–317.
  40. Gold, L.I.; Jussila, T.; Fusenig, N.E.; Stenba, F. TGF-β isoforms are differentially expressed in increasing malignant grades of HaCaT keratinocytes, suggesting separate roles in skin carcinogenesis. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2000, 190, 579–588.
  41. Rodeck, U.; Bossler, A.; Graeven, U.; Fox, F.E.; Nowell, P.C.; Knabbe, C.; Kari, C. Transforming growth factor beta production and responsiveness in normal human melanocytes and melanoma cells. Cancer Res. 1994, 54, 575–581.
  42. van de Vijver, M.J.; He, Y.D.; van ’T Veer, L.; Dai, H.; Hart, A.A.M.; Voskuil, D.W.; Schreiber, G.J.; Peterse, J.L.; Robert, C.; Marton, M.J.; et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 2002, 347, 1999–2009.
  43. van t’ Veer, L.; Dai, H.; van de Vijver, M.J.; He, Y.D.; Hart, A.A.M.; Mao, M.; Peterse, H.L.; van der Koey, K.; Marton, M.J.; Witteveen, A.T.; et al. Gene Expression Profiling Predicts Clinical Outcome of Breast Cancer. Nature 2002, 415, 530–536.
  44. Bueno-de-Mesquita, J.M.; Linn, S.C.; Keijzer, R.; Wesseling, J.; Nuyten, D.S.A.; van Krimpen, C.; Meijers, C.; de Graaf, P.W.; Bos, M.M.E.M.; Hart, A.A.M.; et al. Validation of 70-Gene Prognosis Signature in Node-Negative Breast Cancer. Breast Cancer Res. Treat. 2009, 117, 483–495.
  45. Li, C.; Wang, J.; Wilson, P.B.; Kumar, P.; Levine, E.; Hunter, R.D.; Kumar, S. Role of transforming growth factor β3 in lymphatic metastasis in breast cancer. Int. J. Cancer 1998, 79, 455–459.
  46. Amatschek, S.; Koenig, U.; Auer, H.; Steinlein, P.; Pacher, M.; Gruenfelder, A.; Dekan, G.; Vogl, S.; Kubista, E.; Heider, K.; et al. Tissue-Wide Expression Profiling Using CDNA Subtraction and Microarrays to Identify Tumor-Specific Genes. Cancer Res. 2004, 64, 844–856.
  47. Ghellal, A.; Li, C.; Hayes, M.; Byrne, G.; Bundred, N.; Kumar, S. Prognostic Significance of TGF Beta 1 and TGF Beta 3 in Human Breast Carcinoma. Anticancer Res. 2000, 20, 4413–4418.
  48. Bristow, R.E.; Baldwin, R.L.; Yamada, D.S.; Korc, M.; Karlan, B.Y. Altered Expression of Transforming Growth Factor-Beta Ligands and Receptors in Primary and Recurrent Ovarian Carcinoma. Cancer 1999, 85, 658–668.
  49. Rodriguez, G.C.; Nagarsheth, N.P.; Lee, K.L.; Bentley, R.C.; Walmer, D.K.; Cline, M.; Whitaker, R.S.; Isner, P.; Berchuck, A.; Dodge, R.K.; et al. Progestin-induced apoptosis in the macaque ovarian epithelium: Differential regulation of transforming growth factor-β. J. Natl. Cancer Inst. 2002, 94, 50–60.
  50. Kloen, P.; Gebhardt, M.C.; Perez-atayde, A.; Rosenberg, A.E.; Springfield, D.S.; Gold, L.I.; Ph, D.; Mankin, H.J. Expression of transforming growth factor-β (TGF-β) isoforms in osteosarcomas: TGF-β3 is related to disease progression. Cancer 2000, 80, 2230–2239.
  51. Arici, A.; Sozen, I. Transforming Growth Factor-Β3 Is Expressed at High Levels in Leiomyoma Where It Stimulates Fibronectin Expression and Cell Proliferation. Fertil. Steril. 2000, 73, 1006–1011.
  52. Lee, B.; Nowak, R.A. Human leiomyoma smooth muscle cells show increased expression of transforming growth factor-β3 (TGFβ3) and altered responses to the antiproliferative effects of TGFβ. J. Clin. Endocrinol. Metab. 2001, 86, 913–920.
More
This entry is offline, you can click here to edit this entry!
Video Production Service