The transforming growth factor beta receptors
The transforming growth factor beta (TGF-β) superfamily is a large group of structurally related cell regulatory proteins that was named after its first member, TGF-β1, originally described in 1983.
- Assoian RK, Komoriya A, Meyers CA, Miller DM, Sporn MB (June 1983). “Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization”. J. Biol. Chem. 258 (11): 7155–60. doi:10.1016/S0021-9258(18)32345-7. PMID 6602130.
They interact with TGF-beta receptors.
The transforming growth factor beta (TGFβ) receptors are a family of serine/threonine kinase receptors involved in TGF beta signaling pathway. These receptors bind growth factor and cytokine signaling proteins in the TGF-beta family such as TGFβs (TGFβ1, TGFβ2, TGFβ3), bone morphogenetic proteins (BMPs), growth differentiation factors (GDFs), activin and inhibin, myostatin, anti-Müllerian hormone (AMH), and NODAL. TGFβ family receptors are grouped into three types, type I, type II, and type III. There are seven type I receptors, termed the activin-like receptors (ALK1–7), five type II receptors, and one type III receptor, for a total of 13 TGFβ superfamily receptors. In the transduction pathway, ligand-bound type II receptors activate type I receptors by phosphorylation, which then autophosphorylate and bind SMAD. The Type I receptors have a glycine-serine (GS, or TTSGSGSG) repeat motif of around 30 AA, a target of type II activity. At least three, and perhaps four to five of the serines and threonines in the GS domain, must be phosphorylated to fully activate TbetaR-1.
- “Prosite Documentation PDOC00223”. Archived from the original on 2011-05-25. Retrieved 2006-07-01.
- Fliesler SJ, Kisselev OG (26 December 2007). Signal Transduction in the Retina. CRC Press. pp. 273–. ISBN 978-1-4200-0716-9.
- Thiriet M (14 December 2011). Signaling at the Cell Surface in the Circulatory and Ventilatory Systems. Springer Science & Business Media. pp. 666–. ISBN 978-1-4614-1991-4.
- Wrana JL, Attisano L, Cárcamo J, et al. (December 1992). “TGF beta signals through a heteromeric protein kinase receptor complex”. Cell. 71 (6): 1003–14. doi:10.1016/0092-8674(92)90395-S. PMID 1333888. S2CID 54397586.
- Huse, M; Muir, TW; Xu, L; Chen, YG; Kuriyan, J; Massagué, J (September 2001). “The TGF beta receptor activation process: an inhibitor- to substrate-binding switch”. Molecular Cell. 8 (3): 671–82. doi:10.1016/S1097-2765(01)00332-X. PMID 11583628.
Type I
Type II
Type III
- TGFβR3 (β-glycan)
Unlike the Type I and II receptors which are kinases, TGFBR3 has a Zona pellucida-like domain. Its core domain binds TGF-beta family ligands and its heparan sulfate chains bind bFGF. It acts as a reservoir of ligand for TGF-beta receptors.
- Andres JL, Stanley K, et al. (1989). “Membrane-anchored and soluble forms of betaglycan, a polymorphic proteoglycan that binds transforming growth factor-beta”. J. Cell Biol. 109 (6 (Pt 1)): 3137–3145. doi:10.1083/jcb.109.6.3137. PMC 2115961. PMID 2592419.
- Andres JL, DeFalcis D, et al. (1992). “Binding of two growth factor families to separate domains of the proteoglycan betaglycan”. J. Biol. Chem. 267 (9): 5927–5930. PMID 1556106.
Many proteins have since been described as members of the TGF-β superfamily in a variety of species, including invertebrates as well as vertebrates and categorized into 23 distinct gene types that fall into four major subfamilies.
- Herpin A, Lelong C, Favrel P (May 2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Dev. Comp. Immunol. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
- Burt DW (April 1992). “Evolutionary grouping of the transforming growth factor-beta superfamily”. Biochem. Biophys. Res. Commun. 184 (2): 590–5. doi:10.1016/0006-291X(92)90630-4. PMID 1575734.
- Burt DW, Law AS (1994). “Evolution of the transforming growth factor-beta superfamily” (PDF). Prog. Growth Factor Res. 5 (1): 99–118. doi:10.1016/0955-2235(94)90020-5. hdl:20.500.11820/50fc2d69-c411-4835-9cd9-36bf4144bae4. PMID 8199356. S2CID 41326578.
1. The TGF-β subfamily
Transforming growth factor beta (TGF-β) is a multifunctional cytokine belonging to the transforming growth factor superfamily that includes three different mammalian isoforms (TGF-β 1 to 3, HGNC symbols TGFB1, TGFB2, TGFB3) and many other signaling proteins. TGFB proteins are produced by all white blood cell lineages.
- Meng, Xiao-ming; Nikolic-Paterson, David J.; Lan, Hui Yao (June 2016). “TGF-β: the master regulator of fibrosis”. Nature Reviews Nephrology. 12 (6): 325–338. doi:10.1038/nrneph.2016.48. PMID 27108839. S2CID 25871413.
Activated TGF-β complexes with other factors to form a serine/threonine kinase complex that binds to TGF-β receptors. TGF-β receptors are composed of both type 1 and type 2 receptor subunits. After the binding of TGF-β, the type 2 receptor kinase phosphorylates and activates the type 1 receptor kinase that activates a signaling cascade. This leads to the activation of different downstream substrates and regulatory proteins, inducing transcription of different target genes that function in differentiation, chemotaxis, proliferation, and activation of many immune cells.
- Massagué J (October 2012). “TGFβ signalling in context”. Nature Reviews. Molecular Cell Biology. 13 (10): 616–30. doi:10.1038/nrm3434. PMC 4027049. PMID 22992590.
- Nakao A, Afrakhte M, Morén A, Nakayama T, Christian JL, Heuchel R, et al. (October 1997). “Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling”. Nature. 389 (6651): 631–5. Bibcode:1997Natur.389..631N. doi:10.1038/39369. PMID 9335507. S2CID 4311145.
TGF-β is secreted by many cell types, including macrophages, in a latent form in which it is complexed with two other polypeptides, latent TGF-beta binding protein (LTBP) and latency-associated peptide (LAP). Serum proteinases such as plasmin catalyze the release of active TGF-β from the complex. This often occurs on the surface of macrophages where the latent TGF-β complex is bound to CD36 via its ligand, thrombospondin-1 (TSP-1). Inflammatory stimuli that activate macrophages enhance the release of active TGF-β by promoting the activation of plasmin. Macrophages can also endocytose IgG-bound latent TGF-β complexes that are secreted by plasma cells and then release active TGF-β into the extracellular fluid. Among its key functions is regulation of inflammatory processes, particularly in the gut. TGF-β also plays a crucial role in stem cell differentiation as well as T-cell regulation and differentiation.
- AfCS signaling gateway – data center – ligand description
- Letterio JJ, Roberts AB (April 1998). “Regulation of immune responses by TGF-beta”. Annual Review of Immunology. 16 (1): 137–61. doi:10.1146/annurev.immunol.16.1.137. PMID 9597127.
- Massagué J, Xi Q (July 2012). “TGF-β control of stem cell differentiation genes”. FEBS Letters. 586 (14): 1953–8. doi:10.1016/j.febslet.2012.03.023. PMC 3466472. PMID 22710171.
- Li MO, Flavell RA (August 2008). “TGF-beta: a master of all T cell trades”. Cell. 134 (3): 392–404. doi:10.1016/j.cell.2008.07.025. PMC 3677783. PMID 18692464.
Because of its role in immune and stem cell regulation and differentiation, it is a highly researched cytokine in the fields of cancer, auto-immune diseases, and infectious disease.
The TGF-β superfamily includes endogenous growth inhibiting proteins; an increase in expression of TGF-β often correlates with the malignancy of many cancers and a defect in the cellular growth inhibition response to TGF-β. Its immunosuppressive functions then come to dominate, contributing to oncogenesis.
- Massagué J, Blain SW, Lo RS (October 2000). “TGFbeta signaling in growth control, cancer, and heritable disorders”. Cell. 103 (2): 295–309. doi:10.1016/S0092-8674(00)00121-5. PMID 11057902. S2CID 15482063.
The dysregulation of its immunosuppressive functions is also implicated in the pathogenesis of autoimmune diseases, although their effect is mediated by the environment of other cytokines present.
- Letterio JJ, Roberts AB (April 1998). “Regulation of immune responses by TGF-beta”. Annual Review of Immunology. 16 (1): 137–61. doi:10.1146/annurev.immunol.16.1.137. PMID 9597127.
- Lichtman, Michael K.; Otero-Vinas, Marta; Falanga, Vincent (March 2016). “Transforming growth factor beta (TGF-β) isoforms in wound healing and fibrosis”. Wound Repair and Regeneration. 24 (2): 215–222. doi:10.1111/wrr.12398. ISSN 1524-475X. PMID 26704519. S2CID 4967954.
The primary 3 mammalian types are:
- TGF beta 1 – TGFB1 – Transforming growth factor beta 1 or TGF-β1 is a polypeptide member of the transforming growth factor beta superfamily of cytokines. It is a secreted protein that performs many cellular functions, including the control of cell growth, cell proliferation, cell differentiation, and apoptosis. In humans, TGF-β1 is encoded by the TGFB1 gene. TGF beta 1 has been shown to interact with: Decorin and EIF3I and LTBP and TGF beta receptor 1, and YWHAE.
- Online Mendelian Inheritance in Man (OMIM): 190180
- Universal protein resource accession number P01137 at UniProt.
- Ghadami M, Makita Y, Yoshida K, Nishimura G, Fukushima Y, Wakui K, Ikegawa S, Yamada K, Kondo S, Niikawa N, Tomita Ha (January 2000). “Genetic mapping of the Camurati-Engelmann disease locus to chromosome 19q13.1-q13.3”. Am. J. Hum. Genet. 66 (1): 143–7. doi:10.1086/302728. PMC 1288319. PMID 10631145.
- Vaughn SP, Broussard S, Hall CR, Scott A, Blanton SH, Milunsky JM, Hecht JT (May 2000). “Confirmation of the mapping of the Camurati-Englemann locus to 19q13. 2 and refinement to a 3.2-cM region”. Genomics. 66 (1): 119–21. doi:10.1006/geno.2000.6192. PMID 10843814.
- Hildebrand A, Romarís M, Rasmussen LM, Heinegård D, Twardzik DR, Border WA, Ruoslahti E (September 1994). “Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta”. Biochem. J. 302 (2): 527–34. doi:10.1042/bj3020527. PMC 1137259. PMID 8093006.
- Schönherr E, Broszat M, Brandan E, Bruckner P, Kresse H (July 1998). “Decorin core protein fragment Leu155-Val260 interacts with TGF-beta but does not compete for decorin binding to type I collagen”. Arch. Biochem. Biophys. 355 (2): 241–8. doi:10.1006/abbi.1998.0720. PMID 9675033.
- Takeuchi Y, Kodama Y, Matsumoto T (Dec 1994). “Bone matrix decorin binds transforming growth factor-beta and enhances its bioactivity”. J. Biol. Chem. 269 (51): 32634–8. doi:10.1016/S0021-9258(18)31681-8. PMID 7798269.
- Choy L, Derynck R (November 1998). “The type II transforming growth factor (TGF)-beta receptor-interacting protein TRIP-1 acts as a modulator of the TGF-beta response”. J. Biol. Chem. 273 (47): 31455–62. doi:10.1074/jbc.273.47.31455. PMID 9813058.
- Saharinen J, Keski-Oja J (August 2000). “Specific sequence motif of 8-Cys repeats of TGF-beta binding proteins, LTBPs, creates a hydrophobic interaction surface for binding of small latent TGF-beta”. Mol. Biol. Cell. 11 (8): 2691–704. doi:10.1091/mbc.11.8.2691. PMC 14949. PMID 10930463.
- Ebner R, Chen RH, Lawler S, Zioncheck T, Derynck R (November 1993). “Determination of type I receptor specificity by the type II receptors for TGF-beta or activin”. Science. 262 (5135): 900–2. Bibcode:1993Sci…262..900E. doi:10.1126/science.8235612. PMID 8235612.
- Oh SP, Seki T, Goss KA, Imamura T, Yi Y, Donahoe PK, Li L, Miyazono K, ten Dijke P, Kim S, Li E (March 2000). “Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis”. Proc. Natl. Acad. Sci. U.S.A. 97 (6): 2626–31. Bibcode:2000PNAS…97.2626O. doi:10.1073/pnas.97.6.2626. PMC 15979. PMID 10716993.
- McGonigle S, Beall MJ, Feeney EL, Pearce EJ (February 2001). “Conserved role for 14-3-3epsilon downstream of type I TGFbeta receptors”. FEBS Lett. 490 (1–2): 65–9. doi:10.1016/s0014-5793(01)02133-0. PMID 11172812. S2CID 84710903.
- Further Reading
- Border WA, Noble NA (1994). “Transforming growth factor beta in tissue fibrosis”. N. Engl. J. Med. 331 (19): 1286–92. doi:10.1056/NEJM199411103311907. PMID 7935686.
- Munger JS, Harpel JG, Gleizes PE, Mazzieri R, Nunes I, Rifkin DB (1997). “Latent transforming growth factor-beta: structural features and mechanisms of activation”. Kidney Int. 51 (5): 1376–82. doi:10.1038/ki.1997.188. PMID 9150447.
- Iozzo RV (1999). “The biology of the small leucine-rich proteoglycans. Functional network of interactive proteins”. J. Biol. Chem. 274 (27): 18843–6. doi:10.1074/jbc.274.27.18843. PMID 10383378.
- Reinhold D, Wrenger S, Kähne T, Ansorge S (1999). “HIV-1 Tat: immunosuppression via TGF-beta1 induction”. Immunol. Today. 20 (8): 384–5. doi:10.1016/S0167-5699(99)01497-8. PMID 10431160.
- Yamada Y (2001). “Association of polymorphisms of the transforming growth factor-beta1 gene with genetic susceptibility to osteoporosis”. Pharmacogenetics. 11 (9): 765–71. doi:10.1097/00008571-200112000-00004. PMID 11740340.
- Chen W, Wahl SM (2002). “TGF-β: Receptors, Signaling Pathways and Autoimmunity”. TGF-beta: receptors, signaling pathways and autoimmunity. Curr. Dir. Autoimmun. Current Directions in Autoimmunity. Vol. 5. pp. 62–91. doi:10.1159/000060548. ISBN 978-3-8055-7308-5. PMID 11826761.
- Marone M, Bonanno G, Rutella S, Leone G, Scambia G, Pierelli L (2002). “Survival and cell cycle control in early hematopoiesis: role of bcl-2, and the cyclin dependent kinase inhibitors P27 and P21”. Leuk. Lymphoma. 43 (1): 51–7. doi:10.1080/10428190210195. PMID 11908736. S2CID 28490341.
- Schnaper HW, Hayashida T, Hubchak SC, Poncelet AC (2003). “TGF-beta signal transduction and mesangial cell fibrogenesis”. Am. J. Physiol. Renal Physiol. 284 (2): F243–52. doi:10.1152/ajprenal.00300.2002. PMID 12529270. S2CID 17046094.
- Kalluri R, Neilson EG (2003). “Epithelial-mesenchymal transition and its implications for fibrosis”. J. Clin. Invest. 112 (12): 1776–84. doi:10.1172/JCI20530. PMC 297008. PMID 14679171.
- Grainger DJ (2004). “Transforming growth factor beta and atherosclerosis: so far, so good for the protective cytokine hypothesis”. Arterioscler. Thromb. Vasc. Biol. 24 (3): 399–404. doi:10.1161/01.ATV.0000114567.76772.33. PMID 14699019.
- Attisano L, Labbé E (2004). “TGFbeta and Wnt pathway cross-talk”. Cancer Metastasis Rev. 23 (1–2): 53–61. doi:10.1023/A:1025811012690. PMID 15000149. S2CID 41685620.
- McGowan TA, Zhu Y, Sharma K (2004). “Transforming growth factor-beta: a clinical target for the treatment of diabetic nephropathy”. Curr. Diab. Rep. 4 (6): 447–54. doi:10.1007/s11892-004-0055-z. PMID 15539010. S2CID 45122439.
- Sheppard D (2005). “Integrin-mediated activation of latent transforming growth factor beta”. Cancer Metastasis Rev. 24 (3): 395–402. doi:10.1007/s10555-005-5131-6. PMID 16258727. S2CID 1929903.
- Gressner AM, Weiskirchen R (2006). “Modern pathogenetic concepts of liver fibrosis suggest stellate cells and TGF-beta as major players and therapeutic targets”. J. Cell. Mol. Med. 10 (1): 76–99. doi:10.1111/j.1582-4934.2006.tb00292.x. PMC 3933103. PMID 16563223.
- Seoane J (2006). “Escaping from the TGFbeta anti-proliferative control”. Carcinogenesis. 27 (11): 2148–56. doi:10.1093/carcin/bgl068. PMID 16698802.
- Lee CG, Kang HR, Homer RJ, Chupp G, Elias JA (2006). “Transgenic modeling of transforming growth factor-beta(1): role of apoptosis in fibrosis and alveolar remodeling”. Proc Am Thorac Soc. 3 (5): 418–23. doi:10.1513/pats.200602-017AW. PMC 2658706. PMID 16799085.
- Wahl SM (2007). “Transforming growth factor-beta: innately bipolar”. Curr. Opin. Immunol. 19 (1): 55–62. doi:10.1016/j.coi.2006.11.008. PMID 17137775.
- Redondo S, Santos-Gallego CG, Tejerina T (2007). “TGF-beta1: a novel target for cardiovascular pharmacology”. Cytokine Growth Factor Rev. 18 (3–4): 279–86. doi:10.1016/j.cytogfr.2007.04.005. PMID 17485238.
- Ren H, Han R, Chen X, Liu X, Wan J, Wang L, Yang X, Wang J (May 2020). “Potential therapeutic targets for intracerebral hemorrhage-associated inflammation: An update”. J Cereb Blood Flow Metab. 40 (9): 1752–1768. doi:10.1177/0271678X20923551. PMC 7446569. PMID 32423330. S2CID 218689863.
- TGF beta 2 – TGFB2 – Transforming growth factor-beta 2 (TGF-β2) is a secreted protein known as a cytokine that performs many cellular functions and has a vital role during embryonic development (alternative names: Glioblastoma-derived T-cell suppressor factor, G-TSF, BSC-1 cell growth inhibitor, Polyergin, Cetermin). It is an extracellular glycosylated protein. It is known to suppress the effects of interleukin dependent T-cell tumors. There are two named isoforms of this protein, created by alternative splicing of the same gene (i.e., TGFB2).
- Online Mendelian Inheritance in Man (OMIM): 190220
- Universal protein resource accession number P61812 at UniProt.
- Further Reading
- Clark DA, Coker R (1998). “Transforming growth factor-beta (TGF-beta)”. Int. J. Biochem. Cell Biol. 30 (3): 293–8. doi:10.1016/S1357-2725(97)00128-3. PMID 9611771.
- Wick W, Platten M, Weller M (2002). “Glioma cell invasion: regulation of metalloproteinase activity by TGF-beta”. J. Neurooncol. 53 (2): 177–85. doi:10.1023/A:1012209518843. PMID 11716069. S2CID 21461718.
- Bissell DM (2002). “Chronic liver injury, TGF-beta, and cancer”. Experimental & Molecular Medicine. 33 (4): 179–90. doi:10.1038/emm.2001.31. PMID 11795478.
- Kalluri R, Neilson EG (2004). “Epithelial-mesenchymal transition and its implications for fibrosis”. J. Clin. Invest. 112 (12): 1776–84. doi:10.1172/JCI20530. PMC 297008. PMID 14679171.
- Daopin S, Piez KA, Ogawa Y, Davies DR (1992). “Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily”. Science. 257 (5068): 369–73. Bibcode:1992Sci…257..369D. doi:10.1126/science.1631557. PMID 1631557.
- Schlunegger MP, Grütter MG (1992). “An unusual feature revealed by the crystal structure at 2.2 A resolution of human transforming growth factor-beta 2”. Nature. 358 (6385): 430–4. Bibcode:1992Natur.358..430S. doi:10.1038/358430a0. PMID 1641027. S2CID 4239431.
- Noma T, Glick AB, Geiser AG, et al. (1992). “Molecular cloning and structure of the human transforming growth factor-beta 2 gene promoter”. Growth Factors. 4 (4): 247–55. doi:10.3109/08977199109043910. PMID 1764261.
- Bodmer S, Podlisny MB, Selkoe DJ, et al. (1990). “Transforming growth factor-beta bound to soluble derivatives of the beta amyloid precursor protein of Alzheimer’s disease”. Biochem. Biophys. Res. Commun. 171 (2): 890–7. doi:10.1016/0006-291X(90)91229-L. PMID 2119582.
- Webb NR, Madisen L, Rose TM, Purchio AF (1989). “Structural and sequence analysis of TGF-beta 2 cDNA clones predicts two different precursor proteins produced by alternative mRNA splicing”. DNA. 7 (7): 493–7. doi:10.1089/dna.1.1988.7.493. PMID 2850146.
- Madisen L, Webb NR, Rose TM, et al. (1988). “Transforming growth factor-beta 2: cDNA cloning and sequence analysis”. DNA. 7 (1): 1–8. doi:10.1089/dna.1988.7.1. PMID 3162414.
- Barton DE, Foellmer BE, Du J, et al. (1989). “Chromosomal mapping of genes for transforming growth factors beta 2 and beta 3 in man and mouse: dispersion of TGF-beta gene family”. Oncogene Res. 3 (4): 323–31. PMID 3226728.
- de Martin R, Haendler B, Hofer-Warbinek R, et al. (1988). “Complementary DNA for human glioblastoma-derived T cell suppressor factor, a novel member of the transforming growth factor-beta gene family”. EMBO J. 6 (12): 3673–7. doi:10.1002/j.1460-2075.1987.tb02700.x. PMC 553836. PMID 3322813.
- Marquardt H, Lioubin MN, Ikeda T (1987). “Complete amino acid sequence of human transforming growth factor type beta 2”. J. Biol. Chem. 262 (25): 12127–31. doi:10.1016/S0021-9258(18)45325-2. PMID 3476488.
- Philip A, Bostedt L, Stigbrand T, O’Connor-McCourt MD (1994). “Binding of transforming growth factor-beta (TGF-beta) to pregnancy zone protein (PZP). Comparison to the TGF-beta-alpha 2-macroglobulin interaction”. Eur. J. Biochem. 221 (2): 687–93. doi:10.1111/j.1432-1033.1994.tb18781.x. PMID 7513640.
- Lin HY, Moustakas A, Knaus P, et al. (1995). “The soluble exoplasmic domain of the type II transforming growth factor (TGF)-beta receptor. A heterogeneously glycosylated protein with high affinity and selectivity for TGF-beta ligands”. J. Biol. Chem. 270 (6): 2747–54. doi:10.1074/jbc.270.6.2747. PMID 7852346.
- Hildebrand A, Romarís M, Rasmussen LM, et al. (1994). “Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta”. Biochem. J. 302. ( Pt 2) (2): 527–34. doi:10.1042/bj3020527. PMC 1137259. PMID 8093006.
- López-Casillas F, Payne HM, Andres JL, Massagué J (1994). “Betaglycan can act as a dual modulator of TGF-beta access to signaling receptors: mapping of ligand binding and GAG attachment sites”. J. Cell Biol. 124 (4): 557–68. doi:10.1083/jcb.124.4.557. PMC 2119924. PMID 8106553.
- Fromigué O, Marie PJ, Lomri A (1998). “Bone morphogenetic protein-2 and transforming growth factor-beta2 interact to modulate human bone marrow stromal cell proliferation and differentiation”. J. Cell. Biochem. 68 (4): 411–26. doi:10.1002/(SICI)1097-4644(19980315)68:4<411::AID-JCB2>3.0.CO;2-T. PMID 9493905. S2CID 85850306.
- Mori T, Kawara S, Shinozaki M, et al. (1999). “Role and interaction of connective tissue growth factor with transforming growth factor-beta in persistent fibrosis: A mouse fibrosis model”. J. Cell. Physiol. 181 (1): 153–9. doi:10.1002/(SICI)1097-4652(199910)181:1<153::AID-JCP16>3.0.CO;2-K. PMID 10457363. S2CID 21284888.
- TGF beta 3 – TGFB3 – Transforming growth factor beta-3 is a protein that in humans is encoded by the TGFB3 gene. It is a type of protein, known as a cytokine, which is involved in cell differentiation, embryogenesis and development. It belongs to a large family of cytokines called the Transforming growth factor beta superfamily, which includes the TGF-β family, Bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), inhibins and activins. TGF-β3 is believed to regulate molecules involved in cellular adhesion and extracellular matrix (ECM) formation during the process of palate development. Without TGF-β3, mammals develop a deformity known as a cleft palate. This is caused by failure of epithelial cells in both sides of the developing palate to fuse. TGF-β3 also plays an essential role in controlling the development of lungs in mammals, by also regulating cell adhesion and ECM formation in this tissue, and controls wound healing by regulating the movements of epidermal and dermal cells in injured skin. Transforming growth factor, beta 3 has been shown to interact with TGF beta receptor 2. After successful phase I/II trials, human recombinant TGF-β3 (Avotermin, planned trade name Juvista) failed in Phase III trials.
- Online Mendelian Inheritance in Man (OMIM): 190230
- Universal protein resource accession number P10600 at UniProt.
- Bandyopadhyay B, Fan J, Guan S, Li Y, Chen M, Woodley DT, Li W (Mar 2006). “A “traffic control” role for TGFbeta3: orchestrating dermal and epidermal cell motility during wound healing”. The Journal of Cell Biology. 172 (7): 1093–105. doi:10.1083/jcb.200507111. PMC 2063766. PMID 16549496.
- “Entrez Gene: TGFB3 transforming growth factor, beta 3”.
- Herpin A, Lelong C, Favrel P (May 2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Developmental and Comparative Immunology. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
- Taya Y, O’Kane S, Ferguson MW (Sep 1999). “Pathogenesis of cleft palate in TGF-beta3 knockout mice”. Development. 126 (17): 3869–79. doi:10.1242/dev.126.17.3869. PMID 10433915.
- Dudas M, Nagy A, Laping NJ, Moustakas A, Kaartinen V (Feb 2004). “Tgf-beta3-induced palatal fusion is mediated by Alk-5/Smad pathway”. Developmental Biology. 266 (1): 96–108. doi:10.1016/j.ydbio.2003.10.007. PMID 14729481.
- Kaartinen V, Voncken JW, Shuler C, Warburton D, Bu D, Heisterkamp N, Groffen J (Dec 1995). “Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction”. Nature Genetics. 11 (4): 415–21. doi:10.1038/ng1295-415. PMID 7493022. S2CID 22365206.
- De Crescenzo G, Pham PL, Durocher Y, O’Connor-McCourt MD (May 2003). “Transforming growth factor-beta (TGF-beta) binding to the extracellular domain of the type II TGF-beta receptor: receptor capture on a biosensor surface using a new coiled-coil capture system demonstrates that avidity contributes significantly to high affinity binding”. Journal of Molecular Biology. 328 (5): 1173–83. doi:10.1016/S0022-2836(03)00360-7. PMID 12729750.
- Hart PJ, Deep S, Taylor AB, Shu Z, Hinck CS, Hinck AP (Mar 2002). “Crystal structure of the human TbetaR2 ectodomain–TGF-beta3 complex”. Nature Structural Biology. 9 (3): 203–8. doi:10.1038/nsb766. PMID 11850637. S2CID 13322593.
- Barbara NP, Wrana JL, Letarte M (Jan 1999). “Endoglin is an accessory protein that interacts with the signaling receptor complex of multiple members of the transforming growth factor-beta superfamily”. The Journal of Biological Chemistry. 274 (2): 584–94. doi:10.1074/jbc.274.2.584. PMID 9872992.
- Rotzer D, Roth M, Lutz M, Lindemann D, Sebald W, Knaus P (Feb 2001). “Type III TGF-beta receptor-independent signalling of TGF-beta2 via TbetaRII-B, an alternatively spliced TGF-beta type II receptor”. The EMBO Journal. 20 (3): 480–90. doi:10.1093/emboj/20.3.480. PMC 133482. PMID 11157754.
- Ferguson MW, Duncan J, Bond J, Bush J, Durani P, So K, Taylor L, Chantrey J, Mason T, James G, Laverty H, Occleston NL, Sattar A, Ludlow A, O’Kane S (Apr 2009). “Prophylactic administration of avotermin for improvement of skin scarring: three double-blind, placebo-controlled, phase I/II studies”. Lancet. 373 (9671): 1264–74. doi:10.1016/S0140-6736(09)60322-6. PMID 19362676. S2CID 35671002.
- Renovo shares plummet 75% as scar revision product Juvista fails to meet study endpoints, 14 February 2011
- Further Reading
- Kalluri R, Neilson EG (Dec 2003). “Epithelial-mesenchymal transition and its implications for fibrosis”. The Journal of Clinical Investigation. 112 (12): 1776–84. doi:10.1172/JCI20530. PMC 297008. PMID 14679171.
- Arrick BA, Lee AL, Grendell RL, Derynck R (Sep 1991). “Inhibition of translation of transforming growth factor-beta 3 mRNA by its 5′ untranslated region”. Molecular and Cellular Biology. 11 (9): 4306–13. doi:10.1128/mcb.11.9.4306. PMC 361291. PMID 1875922.
- ten Dijke P, Hansen P, Iwata KK, Pieler C, Foulkes JG (Jul 1988). “Identification of another member of the transforming growth factor type beta gene family”. Proceedings of the National Academy of Sciences of the United States of America. 85 (13): 4715–9. Bibcode:1988PNAS…85.4715T. doi:10.1073/pnas.85.13.4715. PMC 280506. PMID 3164476.
- Derynck R, Lindquist PB, Lee A, Wen D, Tamm J, Graycar JL, Rhee L, Mason AJ, Miller DA, Coffey RJ (Dec 1988). “A new type of transforming growth factor-beta, TGF-beta 3”. The EMBO Journal. 7 (12): 3737–43. doi:10.1002/j.1460-2075.1988.tb03257.x. PMC 454948. PMID 3208746.
- Barton DE, Foellmer BE, Du J, Tamm J, Derynck R, Francke U (1989). “Chromosomal mapping of genes for transforming growth factors beta 2 and beta 3 in man and mouse: dispersion of TGF-beta gene family”. Oncogene Research. 3 (4): 323–31. PMID 3226728.
- Kaartinen V, Voncken JW, Shuler C, Warburton D, Bu D, Heisterkamp N, Groffen J (Dec 1995). “Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction”. Nature Genetics. 11 (4): 415–21. doi:10.1038/ng1295-415. PMID 7493022. S2CID 22365206.
- Nishida K, Sotozono C, Adachi W, Yamamoto S, Yokoi N, Kinoshita S (Mar 1995). “Transforming growth factor-beta 1, -beta 2 and -beta 3 mRNA expression in human cornea”. Current Eye Research. 14 (3): 235–41. doi:10.3109/02713689509033520. PMID 7796607.
- Lin HY, Moustakas A, Knaus P, Wells RG, Henis YI, Lodish HF (Feb 1995). “The soluble exoplasmic domain of the type II transforming growth factor (TGF)-beta receptor. A heterogeneously glycosylated protein with high affinity and selectivity for TGF-beta ligands”. The Journal of Biological Chemistry. 270 (6): 2747–54. doi:10.1074/jbc.270.6.2747. PMID 7852346.
- Rampazzo A, Nava A, Danieli GA, Buja G, Daliento L, Fasoli G, Scognamiglio R, Corrado D, Thiene G (Jun 1994). “The gene for arrhythmogenic right ventricular cardiomyopathy maps to chromosome 14q23-q24”. Human Molecular Genetics. 3 (6): 959–62. doi:10.1093/hmg/3.6.959. PMID 7951245.
- Zhao Y, Chegini N, Flanders KC (Oct 1994). “Human fallopian tube expresses transforming growth factor (TGF beta) isoforms, TGF beta type I-III receptor messenger ribonucleic acid and protein, and contains [125I]TGF beta-binding sites”. The Journal of Clinical Endocrinology and Metabolism. 79 (4): 1177–84. doi:10.1210/jcem.79.4.7962292. PMID 7962292.
- Hildebrand A, Romarís M, Rasmussen LM, Heinegård D, Twardzik DR, Border WA, Ruoslahti E (Sep 1994). “Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta”. The Biochemical Journal. 302 (2): 527–34. doi:10.1042/bj3020527. PMC 1137259. PMID 8093006.
- López-Casillas F, Payne HM, Andres JL, Massagué J (Feb 1994). “Betaglycan can act as a dual modulator of TGF-beta access to signaling receptors: mapping of ligand binding and GAG attachment sites”. The Journal of Cell Biology. 124 (4): 557–68. doi:10.1083/jcb.124.4.557. PMC 2119924. PMID 8106553.
- Mittl PR, Priestle JP, Cox DA, McMaster G, Cerletti N, Grütter MG (Jul 1996). “The crystal structure of TGF-beta 3 and comparison to TGF-beta 2: implications for receptor binding”. Protein Science. 5 (7): 1261–71. doi:10.1002/pro.5560050705. PMC 2143453. PMID 8819159.
- Ambros RA, Kallakury BV, Malfetano JH, Mihm MC (Oct 1996). “Cytokine, cell adhesion receptor, and tumor suppressor gene expression in vulvar squamous carcinoma: correlation with prominent fibromyxoid stromal response”. International Journal of Gynecological Pathology. 15 (4): 320–5. doi:10.1097/00004347-199610000-00004. PMID 8886879.
- Djonov V, Ball RK, Graf S, Mottaz AE, Arnold AM, Flanders K, Studer UE, Merz VW (May 1997). “Transforming growth factor-beta 3 is expressed in nondividing basal epithelial cells in normal human prostate and benign prostatic hyperplasia, and is no longer detectable in prostate carcinoma”. The Prostate. 31 (2): 103–9. doi:10.1002/(SICI)1097-0045(19970501)31:2<103::AID-PROS5>3.0.CO;2-O. PMID 9140123. S2CID 22734222.
- Jin L, Qian X, Kulig E, Sanno N, Scheithauer BW, Kovacs K, Young WF, Lloyd RV (Aug 1997). “Transforming growth factor-beta, transforming growth factor-beta receptor II, and p27Kip1 expression in nontumorous and neoplastic human pituitaries”. The American Journal of Pathology. 151 (2): 509–19. PMC 1858020. PMID 9250163.
- Lidral AC, Romitti PA, Basart AM, Doetschman T, Leysens NJ, Daack-Hirsch S, Semina EV, Johnson LR, Machida J, Burds A, Parnell TJ, Rubenstein JL, Murray JC (Aug 1998). “Association of MSX1 and TGFB3 with nonsyndromic clefting in humans”. American Journal of Human Genetics. 63 (2): 557–68. doi:10.1086/301956. PMC 1377298. PMID 9683588.
- Barbara NP, Wrana JL, Letarte M (Jan 1999). “Endoglin is an accessory protein that interacts with the signaling receptor complex of multiple members of the transforming growth factor-beta superfamily”. The Journal of Biological Chemistry. 274 (2): 584–94. doi:10.1074/jbc.274.2.584. PMID 9872992.
- Lux A, Attisano L, Marchuk DA (Apr 1999). “Assignment of transforming growth factor beta1 and beta3 and a third new ligand to the type I receptor ALK-1”. The Journal of Biological Chemistry. 274 (15): 9984–92. doi:10.1074/jbc.274.15.9984. PMID 10187774.
- Mori T, Kawara S, Shinozaki M, Hayashi N, Kakinuma T, Igarashi A, Takigawa M, Nakanishi T, Takehara K (Oct 1999). “Role and interaction of connective tissue growth factor with transforming growth factor-beta in persistent fibrosis: A mouse fibrosis model”. Journal of Cellular Physiology. 181 (1): 153–9. doi:10.1002/(SICI)1097-4652(199910)181:1<153::AID-JCP16>3.0.CO;2-K. PMID 10457363. S2CID 21284888.
A fourth member, TGF beta 4, has been identified in birds – TGRB4 (synonyms: endometrial bleeding associated factor beta-4 (EBAF)[citation needed], Lefty preproprotein[citation needed], LEFTA[citation needed]; Left-Right Determination Factor 2; LEFTYA; Left-Right Determination Factor A; Transforming Growth Factor Beta-4; Protein Lefty-2; Protein Lefty-A).
- Roberts, Anita B.; Kim, Seong-Jin; Noma, Takafumi; Glick, Adam B.; Lafyatis, Robert; Lechleider, Robert; Jakowlew, Sonia B.; Geiser, Andrew; O’Reilly, Michael A.; Danielpour, David; Sporn, Michael B. (2007). “Multiple Forms of TGF-β: Distinct Promoters and Differential Expression”. Ciba Foundation Symposium 157 – Clinical Applications of TGF-β. Novartis Foundation Symposia. Vol. 157. pp. 7–28. doi:10.1002/9780470514061.ch2. ISBN 978-0-470-51406-1. PMID 1906395.
- Jugessur A, Shi M, Gjessing HK, Lie RT, Wilcox AJ, Weinberg CR, et al. (July 2010). “Maternal genes and facial clefts in offspring: a comprehensive search for genetic associations in two population-based cleft studies from Scandinavia”. PLOS ONE. 5 (7): e11493. Bibcode:2010PLoSO…511493J. doi:10.1371/journal.pone.0011493. PMC 2901336. PMID 20634891.
- He C, Kraft P, Chasman DI, Buring JE, Chen C, Hankinson SE, et al. (November 2010). “A large-scale candidate gene association study of age at menarche and age at natural menopause”. Human Genetics. 128 (5): 515–27. doi:10.1007/s00439-010-0878-4. PMC 2967297. PMID 20734064.
- Otsuki T, Ota T, Nishikawa T, Hayashi K, Suzuki Y, Yamamoto J, et al. (2005). “Signal sequence and keyword trap in silico for selection of full-length human cDNAs encoding secretion or membrane proteins from oligo-capped cDNA libraries”. DNA Research. 12 (2): 117–26. doi:10.1093/dnares/12.2.117. PMID 16303743.
- Cornet PB, Galant C, Eeckhout Y, Courtoy PJ, Marbaix E, Henriet P (February 2005). “Regulation of matrix metalloproteinase-9/gelatinase B expression and activation by ovarian steroids and LEFTY-A/endometrial bleeding-associated factor in the human endometrium”. The Journal of Clinical Endocrinology and Metabolism. 90 (2): 1001–11. doi:10.1210/jc.2004-1277. PMID 15536155.
- Tabibzadeh S, Kothapalli R, Buyuksal I (July 1997). “Distinct tumor specific expression of TGFB4 (ebaf)*, a novel human gene of the TGF-beta superfamily”. Frontiers in Bioscience. 2: a18-25. doi:10.2741/a158. PMID 9230066.
A fifth member of the subfamily, TGFB5, has been identified only in frogs.
- Roberts, Anita B.; Kim, Seong-Jin; Noma, Takafumi; Glick, Adam B.; Lafyatis, Robert; Lechleider, Robert; Jakowlew, Sonia B.; Geiser, Andrew; O’Reilly, Michael A.; Danielpour, David; Sporn, Michael B. (2007). “Multiple Forms of TGF-β: Distinct Promoters and Differential Expression”. Ciba Foundation Symposium 157 – Clinical Applications of TGF-β. Novartis Foundation Symposia. Vol. 157. pp. 7–28. doi:10.1002/9780470514061.ch2. ISBN 978-0-470-51406-1. PMID 1906395.
The peptide structures of the TGF-β isoforms are highly similar (homologies on the order of 70–80%). They are all encoded as large protein precursors; TGF-β1 contains 390 amino acids and TGF-β2 and TGF-β3 each contain 412 amino acids. They each have an N-terminal signal peptide of 20–30 amino acids that they require for secretion from a cell, a pro-region called latency-associated peptide (LAP – Alias: Pro-TGF beta 1, LAP/TGF beta 1), and a 112-114 amino acid C-terminal region that becomes the mature TGF-β molecule following its release from the pro-region by proteolytic cleavage. The mature TGF-β protein dimerizes to produce a 25 KDa active protein with many conserved structural motifs. TGF-β has nine cysteine residues that are conserved among its family. Eight form disulfide bonds within the protein to create a cysteine knot structure characteristic of the TGF-β superfamily. The ninth cysteine forms a disulfide bond with the ninth cysteine of another TGF-β protein to produce a dimer. Many other conserved residues in TGF-β are thought to form secondary structure through hydrophobic interactions. The region between the fifth and sixth conserved cysteines houses the most divergent area of TGF-β proteins that is exposed at the surface of the protein and is implicated in receptor binding and specificity of TGF-β.
- Khalil N (December 1999). “TGF-beta: from latent to active”. Microbes and Infection. 1 (15): 1255–63. doi:10.1016/S1286-4579(99)00259-2. PMID 10611753.
- Herpin A, Lelong C, Favrel P (May 2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Developmental and Comparative Immunology. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
- Daopin S, Piez KA, Ogawa Y, Davies DR (July 1992). “Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily”. Science. 257 (5068): 369–73. Bibcode:1992Sci…257..369D. doi:10.1126/science.1631557. PMID 1631557.
Although TGF-β is important in regulating crucial cellular activities, only a few TGF-β activating pathways are currently known, and the full mechanism behind the suggested activation pathways is not yet well understood. Some of the known activating pathways are cell or tissue specific, while some are seen in multiple cell types and tissues. Proteases, integrins, pH, and reactive oxygen species are just few of the currently known factors that can activate TGF-β, as discussed below. It is well known that perturbations of these activating factors can lead to unregulated TGF-β signaling levels that may cause several complications including inflammation, autoimmune disorders, fibrosis, cancer and cataracts. In most cases, an activated TGF-β ligand will initiate the TGF-β signaling cascade as long as TGF-β receptors I and II are available for binding. This is due to a high affinity between TGF-β and its receptors, suggesting why the TGF-β signaling recruits a latency system to mediate its signaling.
- Annes JP, Munger JS, Rifkin DB (January 2003). “Making sense of latent TGFbeta activation”. Journal of Cell Science. 116 (Pt 2): 217–24. doi:10.1242/jcs.00229. PMID 12482908.
- ten Dijke P, Hill CS (May 2004). “New insights into TGF-beta-Smad signalling”. Trends in Biochemical Sciences. 29 (5): 265–73. doi:10.1016/j.tibs.2004.03.008. PMID 15130563.
- Stetler-Stevenson WG, Aznavoorian S, Liotta LA (1993). “Tumor cell interactions with the extracellular matrix during invasion and metastasis”. Annual Review of Cell Biology. 9: 541–73. doi:10.1146/annurev.cb.09.110193.002545. PMID 8280471.
- Barcellos-Hoff MH, Dix TA (September 1996). “Redox-mediated activation of latent transforming growth factor-beta 1”. Molecular Endocrinology. 10 (9): 1077–83. doi:10.1210/mend.10.9.8885242. PMID 8885242.
- Wipff PJ, Hinz B (September 2008). “Integrins and the activation of latent transforming growth factor beta1 – an intimate relationship”. European Journal of Cell Biology. 87 (8–9): 601–15. doi:10.1016/j.ejcb.2008.01.012. PMID 18342983.
- Yu Q, Stamenkovic I (January 2000). “Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis”. Genes & Development. 14 (2): 163–76. doi:10.1101/gad.14.2.163. PMC 316345. PMID 10652271.
- Taipale J, Miyazono K, Heldin CH, Keski-Oja J (January 1994). “Latent transforming growth factor-beta 1 associates to fibroblast extracellular matrix via latent TGF-beta binding protein”. The Journal of Cell Biology. 124 (1–2): 171–81. doi:10.1083/jcb.124.1.171. PMC 2119892. PMID 8294500.
Integrin-independent activation
Activation by protease and metalloprotease
Plasmin and a number of matrix metalloproteinases (MMP) play a key role in promoting tumor invasion and tissue remodeling by inducing proteolysis of several ECM components. The TGF-β activation process involves the release of the LLC from the matrix, followed by further proteolysis of the LAP to release TGF-β to its receptors. MMP-9 and MMP-2 are known to cleave latent TGF-β. The LAP complex contains a protease-sensitive hinge region which can be the potential target for this liberation of TGF-β. Despite the fact that MMPs have been proven to play a key role in activating TGF-β, mice with mutations in MMP-9 and MMP-2 genes can still activate TGF-β and do not show any TGF-β deficiency phenotypes, this may reflect redundancy among the activating enzymes suggesting that other unknown proteases might be involved.
- Stetler-Stevenson WG, Aznavoorian S, Liotta LA (1993). “Tumor cell interactions with the extracellular matrix during invasion and metastasis”. Annual Review of Cell Biology. 9: 541–73.
- Yu Q, Stamenkovic I (January 2000). “Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis”. Genes & Development. 14 (2): 163–76. doi:10.1101/gad.14.2.163. PMC 316345. PMID 10652271.
- Taipale J, Miyazono K, Heldin CH, Keski-Oja J (January 1994). “Latent transforming growth factor-beta 1 associates to fibroblast extracellular matrix via latent TGF-beta binding protein”. The Journal of Cell Biology. 124 (1–2): 171–81. doi:10.1083/jcb.124.1.171. PMC 2119892. PMID 8294500.
- Annes JP, Munger JS, Rifkin DB (January 2003). “Making sense of latent TGFbeta activation”. Journal of Cell Science. 116 (Pt 2): 217–24. doi:10.1242/jcs.00229. PMID 12482908.
Activation by pH
Acidic conditions can denature the LAP. Treatment of the medium with extremes of pH (1.5 or 12) resulted in significant activation of TGF-β as shown by radio-receptor assays, while mild acid treatment (pH 4.5) yielded only 20-30% of the activation achieved by pH 1.5.
Average pH of common solutions | ||
---|---|---|
Substance | pH range | Type |
Battery acid | < 1 | Acid |
Gastric acid | 1.0 – 1.5 | |
Vinegar | 4-5 | |
Orange juice | 3.3 – 4.2 | |
Black coffee | 5 – 5.03 | |
Milk | 6.5 – 6.8 | |
Pure water at 25 °C | 7 | Neutral |
Sea water | 7.5 – 8.4 | Base |
Ammonia | 11.0 – 11.5 | |
Bleach | 12.5 | |
1 M NaOH | 14 |
- Lyons RM, Keski-Oja J, Moses HL (May 1988). “Proteolytic activation of latent transforming growth factor-beta from fibroblast-conditioned medium”. The Journal of Cell Biology. 106 (5): 1659–65. doi:10.1083/jcb.106.5.1659. PMC 2115066. PMID 2967299.
Activation by reactive oxygen species (ROS)
The structure of LAP is important in maintaining its function. Structure modification of LAP can lead to disturb the interaction between LAP and TGF-β and thus activating it. Factors that may cause such modification may include hydroxyl radicals from reactive oxygen species (ROS). TGF-β was rapidly activated after in vivo radiation exposure ROS.
- Barcellos-Hoff MH, Dix TA (September 1996). “Redox-mediated activation of latent transforming growth factor-beta 1”. Molecular Endocrinology. 10 (9): 1077–83. doi:10.1210/mend.10.9.8885242. PMID 8885242.
Activation by thrombospondin-1
Thrombospondin-1 (TSP-1) is a matricellular glycoprotein found in plasma of healthy patients with levels in the range of 50–250 ng/ml. TSP-1 levels are known to increase in response to injury and during development. TSP-1 activates latent TGF-beta by forming direct interactions with the latent TGF-β complex and induces a conformational rearrangement preventing it from binding to the matured TGF-β.
- Booth WJ, Berndt MC (July 1987). “Thrombospondin in clinical disease states”. Seminars in Thrombosis and Hemostasis. 13 (3): 298–306. doi:10.1055/s-2007-1003505. PMID 3317840.
- Raugi GJ, Olerud JE, Gown AM (December 1987). “Thrombospondin in early human wound tissue”. The Journal of Investigative Dermatology. 89 (6): 551–4. doi:10.1111/1523-1747.ep12461198. PMID 3680981.
- Schultz-Cherry S, Murphy-Ullrich JE (August 1993). “Thrombospondin causes activation of latent transforming growth factor-beta secreted by endothelial cells by a novel mechanism”. The Journal of Cell Biology. 122 (4): 923–32. doi:10.1083/jcb.122.4.923. PMC 2119591. PMID 8349738.
- Murphy-Ullrich JE, Poczatek M (2000). “Activation of latent TGF-beta by thrombospondin-1: mechanisms and physiology”. Cytokine & Growth Factor Reviews. 11 (1–2): 59–69. doi:10.1016/S1359-6101(99)00029-5. PMID 10708953.
Activation by Alpha(V) containing integrins
The general theme of integrins participating in latent TGF-β1 activation arose from studies that examined mutations/knockouts of β6 integrin, αV integrin, β8 integrin and in LAP. These mutations produced phenotypes that were similar to phenotypes seen in TGF-β1 knockout mice. Currently there are two proposed models of how αV containing integrins can activate latent TGF-β1; the first proposed model is by inducing conformational change to the latent TGF-β1 complex and hence releasing the active TGF-β1 and the second model is by a protease-dependent mechanism.
- Huang XZ, Wu JF, Cass D, Erle DJ, Corry D, Young SG, et al. (May 1996). “Inactivation of the integrin beta 6 subunit gene reveals a role of epithelial integrins in regulating inflammation in the lung and skin”. The Journal of Cell Biology. 133 (4): 921–8. doi:10.1083/jcb.133.4.921. PMC 2120829. PMID 8666675.
- Bader BL, Rayburn H, Crowley D, Hynes RO (November 1998). “Extensive vasculogenesis, angiogenesis, and organogenesis precede lethality in mice lacking all alpha v integrins”. Cell. 95 (4): 507–19. doi:10.1016/S0092-8674(00)81618-9. PMID 9827803. S2CID 13076974.
- Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, et al. (October 1992). “Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease”. Nature. 359 (6397): 693–9. Bibcode:1992Natur.359..693S. doi:10.1038/359693a0. PMC 3889166. PMID 1436033.
- Wipff PJ, Hinz B (September 2008). “Integrins and the activation of latent transforming growth factor beta1 – an intimate relationship”. European Journal of Cell Biology. 87 (8–9): 601–15. doi:10.1016/j.ejcb.2008.01.012. PMID 18342983.
Conformation change mechanism pathway (without proteolysis)
αVβ6 integrin was the first integrin to be identified as TGF-β1 activator. LAPs contain an RGD motif which is recognized by vast majority of αV containing integrins, and αVβ6 integrin can activate TGF-β1 by binding to the RGD motif present in LAP-β1 and LAP-β3. Upon binding, it induces adhesion-mediated cell forces that are translated into biochemical signals which can lead to liberation/activation of TGFb from its latent complex. This pathway has been demonstrated for activation of TGF-β in epithelial cells and does not associate MMPs.
- Annes JP, Munger JS, Rifkin DB (January 2003). “Making sense of latent TGFbeta activation”. Journal of Cell Science. 116 (Pt 2): 217–24. doi:10.1242/jcs.00229. PMID 12482908.
- Munger JS, Harpel JG, Giancotti FG, Rifkin DB (September 1998). “Interactions between growth factors and integrins: latent forms of transforming growth factor-beta are ligands for the integrin alphavbeta1”. Molecular Biology of the Cell. 9 (9): 2627–38. CiteSeerX 10.1.1.492.5809. doi:10.1091/mbc.9.9.2627. PMC 25536. PMID 9725916.
- Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J, et al. (February 1999). “The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis”. Cell. 96 (3): 319–28. doi:10.1016/S0092-8674(00)80545-0. PMID 10025398.
- Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, Flanders KC, et al. (January 1993). “Transforming growth factor beta 1 null mutation in mice causes excessive inflammatory response and early death”. Proceedings of the National Academy of Sciences of the United States of America. 90 (2): 770–4. Bibcode:1993PNAS…90..770K. doi:10.1073/pnas.90.2.770. PMC 45747. PMID 8421714.
- Taylor AW (January 2009). “Review of the activation of TGF-beta in immunity”. Journal of Leukocyte Biology. 85 (1): 29–33. doi:10.1189/jlb.0708415. PMC 3188956. PMID 18818372.
Integrin protease-dependent activation mechanism
Because MMP-2 and MMP-9 can activate TGF-β through proteolytic degradation of the latent TGF beta complex, αV containing integrins activate TGF-β1 by creating a close connection between the latent TGF-β complex and MMPs. Integrins αVβ6 and αVβ3 are suggested to simultaneously bind the latent TGF-β1 complex and proteinases, simultaneous inducing conformational changes of the LAP and sequestering proteases to close proximity. Regardless of involving MMPs, this mechanism still necessitate the association of integrins and that makes it a non proteolytic pathway.
- Wipff PJ, Hinz B (September 2008). “Integrins and the activation of latent transforming growth factor beta1 – an intimate relationship”. European Journal of Cell Biology. 87 (8–9): 601–15. doi:10.1016/j.ejcb.2008.01.012. PMID 18342983.
- Yu Q, Stamenkovic I (January 2000). “Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis”. Genes & Development. 14 (2): 163–76. doi:10.1101/gad.14.2.163. PMC 316345. PMID 10652271.
- Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, et al. (April 2002). “The integrin alpha(v)beta8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-beta1”. The Journal of Cell Biology. 157 (3): 493–507. doi:10.1083/jcb.200109100. PMC 2173277. PMID 11970960.
Further reading
- Moses HL, Roberts AB, Derynck R (July 2016). “The Discovery and Early Days of TGF-β: A Historical Perspective”. Cold Spring Harbor Perspectives in Biology. 8 (7): a021865. doi:10.1101/cshperspect.a021865. PMC 4930926. PMID 27328871.
- Derynck R, Zhang Y, Feng XH (December 1998). “Smads: transcriptional activators of TGF-beta responses”. Cell. 95 (6): 737–40. doi:10.1016/S0092-8674(00)81696-7. PMID 9865691. S2CID 17711163.
- Derynck R, Zhang YE (October 2003). “Smad-dependent and Smad-independent pathways in TGF-beta family signalling”. Nature. 425 (6958): 577–84. Bibcode:2003Natur.425..577D. doi:10.1038/nature02006. PMID 14534577. S2CID 4419607
- Letterio JJ (August 2005). “TGF-beta signaling in T cells: roles in lymphoid and epithelial neoplasia”. Oncogene. 24 (37): 5701–12. doi:10.1038/sj.onc.1208922. PMID 16123803.
- Zhang YE (February 2017). “Non-Smad Signaling Pathways of the TGF-β Family”. Cold Spring Harbor Perspectives in Biology. 9 (2): a022129. doi:10.1101/cshperspect.a022129. PMC 5287080. PMID 27864313.
- Roskoski R (August 2012). “ERK1/2 MAP kinases: structure, function, and regulation”. Pharmacological Research. 66 (2): 105–43. doi:10.1016/j.phrs.2012.04.005. PMID 22569528.
- Chen IT, Hsu PH, Hsu WC, Chen NJ, Tseng PH (July 2015). “Polyubiquitination of Transforming Growth Factor β-activated Kinase 1 (TAK1) at Lysine 562 Residue Regulates TLR4-mediated JNK and p38 MAPK Activation”. Scientific Reports. 5: 12300. Bibcode:2015NatSR…512300C. doi:10.1038/srep12300. PMC 4507259. PMID 26189595.
- Yang X, Khosravi-Far R, Chang HY, Baltimore D (June 1997). “Daxx, a novel Fas-binding protein that activates JNK and apoptosis”. Cell. 89 (7): 1067–76. doi:10.1016/S0092-8674(00)80294-9. PMC 2989411. PMID 9215629.
- Perlman R, Schiemann WP, Brooks MW, Lodish HF, Weinberg RA (August 2001). “TGF-beta-induced apoptosis is mediated by the adapter protein Daxx that facilitates JNK activation”. Nature Cell Biology. 3 (8): 708–14. doi:10.1038/35087019. PMID 11483955. S2CID 20435808.
- Hofmann TG, Stollberg N, Schmitz ML, Will H (December 2003). “HIPK2 regulates transforming growth factor-beta-induced c-Jun NH(2)-terminal kinase activation and apoptosis in human hepatoma cells”. Cancer Research. 63 (23): 8271–7. PMID 14678985.
- Tu, Wan-zhi; Fu, Yan-bin; Xie, Xin (December 2019). “RepSox, a small molecule inhibitor of the TGFβ receptor, induces brown adipogenesis and browning of white adipocytes”. Acta Pharmacologica Sinica. 40 (12): 1523–1531. doi:10.1038/s41401-019-0264-2. PMC 7471457. PMID 31235818.
- Inman, Gareth J.; Nicolás, Francisco J.; Callahan, James F.; Harling, John D.; Gaster, Laramie M.; Reith, Alastair D.; Laping, Nicholas J.; Hill, Caroline S. (1 July 2002). “SB-431542 Is a Potent and Specific Inhibitor of Transforming Growth Factor-β Superfamily Type I Activin Receptor-Like Kinase (ALK) Receptors ALK4, ALK5, and ALK7”. Molecular Pharmacology. 62 (1): 65–74. doi:10.1124/mol.62.1.65. PMID 12065756. S2CID 15185199.
- Tojo, Masayoshi; Hamashima, Yoshio; Hanyu, Aki; Kajimoto, Tetsuya; Saitoh, Masao; Miyazono, Kohei; Node, Manabu; Imamura, Takeshi (November 2005). “The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-beta”. Cancer Science. 96 (11): 791–800. doi:10.1111/j.1349-7006.2005.00103.x. PMID 16271073. S2CID 3105729.
- Yingling, Jonathan M.; McMillen, William T.; Yan, Lei; Huang, Huocong; Sawyer, J. Scott; Graff, Jeremy; Clawson, David K.; Britt, Karen S.; Anderson, Bryan D.; Beight, Douglas W.; Desaiah, Durisala; Lahn, Michael M.; Benhadji, Karim A.; Lallena, Maria J.; Holmgaard, Rikke B.; Xu, Xiaohong; Zhang, Faming; Manro, Jason R.; Iversen, Philip W.; Iyer, Chandrasekar V.; Brekken, Rolf A.; Kalos, Michael D.; Driscoll, Kyla E. (23 January 2018). “Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor”. Oncotarget. 9 (6): 6659–6677. doi:10.18632/oncotarget.23795. PMC 5805504. PMID 29467918.
- Johnston, Chris J. C.; Smyth, Danielle J.; Kodali, Ravindra B.; White, Madeleine P. J.; Harcus, Yvonne; Filbey, Kara J.; Hewitson, James P.; Hinck, Cynthia S.; Ivens, Alasdair; Kemter, Andrea M.; Kildemoes, Anna O.; Le Bihan, Thierry; Soares, Dinesh C.; Anderton, Stephen M.; Brenn, Thomas; Wigmore, Stephen J.; Woodcock, Hannah V.; Chambers, Rachel C.; Hinck, Andrew P.; McSorley, Henry J.; Maizels, Rick M. (December 2017). “A structurally distinct TGF-β mimic from an intestinal helminth parasite potently induces regulatory T cells”. Nature Communications. 8 (1): 1741. Bibcode:2017NatCo…8.1741J. doi:10.1038/s41467-017-01886-6. PMC 5701006. PMID 29170498.
- Smyth, Danielle J.; Harcus, Yvonne; White, Madeleine P.J.; Gregory, William F.; Nahler, Janina; Stephens, Ian; Toke-Bjolgerud, Edward; Hewitson, James P.; Ivens, Alasdair; McSorley, Henry J.; Maizels, Rick M. (April 2018). “TGF-β mimic proteins form an extended gene family in the murine parasite Heligmosomoides polygyrus”. International Journal for Parasitology. 48 (5): 379–385. doi:10.1016/j.ijpara.2017.12.004. PMC 5904571. PMID 29510118.
- Mukundan, Ananya; Byeon, Chang-Hyeock; Hinck, Cynthia S.; Smyth, Danielle J.; Maizels, Rick M.; Hinck, Andrew P. (2020-12-09). “Structure-based mapping of the TβRI and TβRII receptor binding sites of the parasitic TGF-β mimic, Hp-TGM”. doi:10.1101/2020.12.08.416701. S2CID 229293860.
- Cook, Laura; Reid, Kyle T; Häkkinen, Elmeri; de Bie, Brett; Tanaka, Shigeru; Smyth, Danielle J; White, Madeleine PJ; Wong, May Q; Huang, Qing; Gillies, Jana K; Zeigler, Steven F; Maizels, Rick M; Levings, Megan K (2021-04-30). “Induction of stable human FOXP3+ Tregs by a parasite-derived TGF-β mimic”. Immunology & Cell Biology. 99 (8): 833–847. doi:10.1111/imcb.12475. PMC 8453874. PMID 33929751.
- Eisenstein EM, Williams CB (May 2009). “The T(reg)/Th17 cell balance: a new paradigm for autoimmunity”. Pediatric Research. 65 (5 Pt 2): 26R–31R. doi:10.1203/PDR.0b013e31819e76c7. PMID 19218879. S2CID 1778541.
- Morishima N, Mizoguchi I, Takeda K, Mizuguchi J, Yoshimoto T (August 2009). “TGF-beta is necessary for induction of IL-23R and Th17 differentiation by IL-6 and IL-23”. Biochemical and Biophysical Research Communications. 386 (1): 105–10. doi:10.1016/j.bbrc.2009.05.140. PMID 19501566.
- Li MO, Flavell RA (August 2008). “TGF-beta: a master of all T cell trades”. Cell. 134 (3): 392–404. doi:10.1016/j.cell.2008.07.025. PMC 3677783. PMID 18692464.
- O’Connor W, Zenewicz LA, Flavell RA (June 2010). “The dual nature of T(H)17 cells: shifting the focus to function”. Nature Immunology. 11 (6): 471–6. doi:10.1038/ni.1882. PMID 20485275. S2CID 2980102.
- Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA (2006-01-01). “Transforming growth factor-beta regulation of immune responses”. Annual Review of Immunology. 24 (1): 99–146. doi:10.1146/annurev.immunol.24.021605.090737. PMID 16551245.
- Roes J, Choi BK, Cazac BB (June 2003). “Redirection of B cell responsiveness by transforming growth factor beta receptor”. Proceedings of the National Academy of Sciences of the United States of America. 100 (12): 7241–6. Bibcode:2003PNAS..100.7241R. doi:10.1073/pnas.0731875100. PMC 165860. PMID 12773615.
- Patil S, Wildey GM, Brown TL, Choy L, Derynck R, Howe PH (December 2000). “Smad7 is induced by CD40 and protects WEHI 231 B-lymphocytes from transforming growth factor-beta -induced growth inhibition and apoptosis”. The Journal of Biological Chemistry. 275 (49): 38363–70. doi:10.1074/jbc.M004861200. PMID 10995749.
- Arsura M, Wu M, Sonenshein GE (July 1996). “TGF beta 1 inhibits NF-kappa B/Rel activity inducing apoptosis of B cells: transcriptional activation of I kappa B alpha”. Immunity. 5 (1): 31–40. doi:10.1016/S1074-7613(00)80307-6. PMID 8758892.
- Kubiczkova L, Sedlarikova L, Hajek R, Sevcikova S (September 2012). “TGF-β – an excellent servant but a bad master”. Journal of Translational Medicine. 10 (1): 183. doi:10.1186/1479-5876-10-183. PMC 3494542. PMID 22943793.
- Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, et al. (January 2005). “Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity”. The Journal of Clinical Investigation. 115 (1): 66–75. doi:10.1172/JCI19229. PMC 539188. PMID 15630445.
- Wahl SM (February 2007). “Transforming growth factor-beta: innately bipolar”. Current Opinion in Immunology. 19 (1): 55–62. doi:10.1016/j.coi.2006.11.008. PMID 17137775.
- Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, et al. (April 2011). “Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis”. Science. 332 (6026): 243–7. Bibcode:2011Sci…332..243W. doi:10.1126/science.1201475. PMC 3144160. PMID 21436399
- Hanahan D, Weinberg RA (January 2000). “The hallmarks of cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9. PMID 10647931. S2CID 1478778.
- Blobe GC, Schiemann WP, Lodish HF (May 2000). “Role of transforming growth factor beta in human disease”. The New England Journal of Medicine. 342 (18): 1350–8. doi:10.1056/NEJM200005043421807. PMID 10793168.
- Hassan, Sobia; Shah, Hussain; Shawana, Summayya (2020). “Dysregulated epidermal growth factor and tumor growth factor-beta receptor signaling through GFAP-ACTA2 protein interaction in liver fibrosis”. Pakistan Journal of Medical Sciences. 36 (4): 782–787. doi:10.12669/pjms.36.4.1845. PMC 7260937. PMID 32494274.
- Vlahopoulos SA, Cen O, Hengen N, Agan J, Moschovi M, Critselis E, et al. (August 2015). “Dynamic aberrant NF-κB spurs tumorigenesis: a new model encompassing the microenvironment”. Cytokine & Growth Factor Reviews. 26 (4): 389–403. doi:10.1016/j.cytogfr.2015.06.001. PMC 4526340. PMID 26119834.
- Ezzeddini R, Somi MH, Taghikhani M, Moaddab SY, Masnadi Shirazi K, Shirmohammadi M, Eftekharsadat AT, Sadighi Moghaddam B, Salek Farrokhi A (February 2021). “Association of Foxp3 rs3761548 polymorphism with cytokines concentration in gastric adenocarcinoma patients”. Cytokine. 138: 155351. doi:10.1016/j.cyto.2020.155351. ISSN 1043-4666. PMID 33127257. S2CID 226218796.
- Bonecini-Almeida, M. Glória; Ho, John L.; Boéchat, Neio; Huard, Richard C.; Chitale, Sadhana; Doo, Howard; Geng, Jiayuan; Rego, Lorena; Lazzarini, Luiz Claudio Oliveira; Kritski, Afrânio L.; Johnson, Warren D.; McCaffrey, Timothy A.; Silva, José R. Lapa e (May 2004). “Down-Modulation of Lung Immune Responses by Interleukin-10 and Transforming Growth Factor β (TGF-β) and Analysis of TGF-β Receptors I and II in Active Tuberculosis”. Infection and Immunity. 72 (5): 2628–2634. doi:10.1128/IAI.72.5.2628-2634.2004. PMC 387880. PMID 15102771.
- Rook, Graham A. W.; Lowrie, Douglas B.; Hernàndez-Pando, Rogelio (15 July 2007). “Immunotherapeutics for Tuberculosis in Experimental Animals: Is There a Common Pathway Activated by Effective Protocols?”. The Journal of Infectious Diseases. 196 (2): 191–198. doi:10.1086/518937. PMID 17570105.
- Warsinske, Hayley C.; Pienaar, Elsje; Linderman, Jennifer J.; Mattila, Joshua T.; Kirschner, Denise E. (20 December 2017). “Deletion of TGF-β1 Increases Bacterial Clearance by Cytotoxic T Cells in a Tuberculosis Granuloma Model”. Frontiers in Immunology. 8: 1843. doi:10.3389/fimmu.2017.01843. PMC 5742530. PMID 29326718.
- Gern, Benjamin H.; Adams, Kristin N.; Plumlee, Courtney R.; Stoltzfus, Caleb R.; Shehata, Laila; Moguche, Albanus O.; Busman-Sahay, Kathleen; Hansen, Scott G.; Axthelm, Michael K.; Picker, Louis J.; Estes, Jacob D.; Urdahl, Kevin B.; Gerner, Michael Y. (March 2021). “TGFβ restricts expansion, survival, and function of T cells within the tuberculous granuloma”. Cell Host & Microbe. 29 (4): 594–606.e6. doi:10.1016/j.chom.2021.02.005. PMC 8624870. PMID 33711270. S2CID 232217715.
- Jayaswal, Shilpi; Kamal, Md. Azhar; Dua, Raina; Gupta, Shashank; Majumdar, Tanmay; Das, Gobardhan; Kumar, Dhiraj; Rao, Kanury V. S. (15 April 2010). “Identification of Host-Dependent Survival Factors for Intracellular Mycobacterium tuberculosis through an siRNA Screen”. PLOS Pathogens. 6 (4): e1000839. doi:10.1371/journal.ppat.1000839. PMC 2855445. PMID 20419122. S2CID 1769658
- Rosas-Taraco, Adrian G.; Higgins, David M.; Sánchez-Campillo, Joaquín; Lee, Eric J.; Orme, Ian M.; González-Juarrero, Mercedes (January 2011). “Local pulmonary immunotherapy with siRNA targeting TGFβ1 enhances antimicrobial capacity in Mycobacterium tuberculosis infected mice”. Tuberculosis. 91 (1): 98–106. doi:10.1016/j.tube.2010.11.004. PMC 7238550. PMID 21195670.
- Understanding Heart Disease: Research Explains Link Between Cholesterol and Heart Disease Archived 2007-11-12 at the Wayback Machine
- Entrez Gene (2007). “TGFBR2 transforming growth factor, beta receptor II” (Entrez gene entry). Retrieved January 11, 2007.
- Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK, et al. (April 2006). “Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome”. Science. 312 (5770): 117–21. Bibcode:2006Sci…312..117H. doi:10.1126/science.1124287. PMC 1482474. PMID 16601194.
- Robinson PN, Arteaga-Solis E, Baldock C, Collod-Béroud G, Booms P, De Paepe A, et al. (October 2006). “The molecular genetics of Marfan syndrome and related disorders”. Journal of Medical Genetics. 43 (10): 769–87. doi:10.1136/jmg.2005.039669. PMC 2563177. PMID 16571647.
- Selvaraj RK, Geiger TL (March 2008). “Mitigation of experimental allergic encephalomyelitis by TGF-β induced FOXP3+ regulatory T cells through the induction of anergy and infectious tolerance”. Journal of Immunology. 180 (5): 2830–8. doi:10.4049/jimmunol.180.5.2830. PMID 18292504.
- Dobolyi A, Vincze C, Pál G, Lovas G (July 2012). “The neuroprotective functions of transforming growth factor beta proteins”. International Journal of Molecular Sciences. 13 (7): 8219–58. doi:10.3390/ijms13078219. PMC 3430231. PMID 22942700.
- Nakahara J, Maeda M, Aiso S, Suzuki N (February 2012). “Current concepts in multiple sclerosis: autoimmunity versus oligodendrogliopathy”. Clinical Reviews in Allergy & Immunology. 42 (1): 26–34. doi:10.1007/s12016-011-8287-6. PMID 22189514. S2CID 21058811.
- Swardfager W, Lanctôt K, Rothenburg L, Wong A, Cappell J, Herrmann N (November 2010). “A meta-analysis of cytokines in Alzheimer’s disease”. Biological Psychiatry. 68 (10): 930–41. doi:10.1016/j.biopsych.2010.06.012. PMID 20692646. S2CID 6544784.
- Senatorov VV, Friedman, AR, Milikovsky DZ, Ofer J, Saar-Ashkenazy R, Charbash A, Jahan N, Chin G, Mihaly E, Lin JM, Ramsay HJ, Moghbel A, Preininger MK, Eddings CR, Harrison HV, Patel R, Shen Y, Ghanim H, Sheng H, Veksler R, Sudmant PH, Becker A, Hart B, Rogawski MA, Dillin A, Friedman A, and Kaufer D (December 2019). “Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction”. Science Translational Medicine. 11 (521): eaaw8283. bioRxiv 10.1101/537431. doi:10.1126/scitranslmed.aaw8283. PMID 31801886.
- Daniela Kaufer; Alon Friedman (May 2021). “Damage to a Protective Shield around the Brain May Lead to Alzheimer’s and Other Diseases”. Scientific American.
- Maier P, Broszinski A, Heizmann U, Böhringer D, Reinhardau T (July 2007). “Active transforming growth factor-beta2 is increased in the aqueous humor of keratoconus patients”. Molecular Vision. 13: 1198–202. PMID 17679942.
- Engler C, Chakravarti S, Doyle J, Eberhart CG, Meng H, Stark WJ, et al. (May 2011). “Transforming growth factor-β signaling pathway activation in Keratoconus”. American Journal of Ophthalmology. 151 (5): 752–759.e2. doi:10.1016/j.ajo.2010.11.008. PMC 3079764. PMID 21310385.
- Oddy WH, Rosales F (February 2010). “A systematic review of the importance of milk TGF-beta on immunological outcomes in the infant and young child”. Pediatric Allergy and Immunology. 21 (1 Pt 1): 47–59. doi:10.1111/j.1399-3038.2009.00913.x. PMID 19594862.
- Galindo, Yadira (2018-12-26). “UC San Diego Researchers Identify How Skin Ages, Loses Fat and Immunity” (Press release). University of California San Diego.
- Zhang LJ, Chen SX, Guerrero-Juarez CF, Li F, Tong Y, Liang Y, et al. (January 2019). “Age-Related Loss of Innate Immune Antimicrobial Function of Dermal Fat Is Mediated by Transforming Growth Factor Beta”. Immunity. 50 (1): 121–136.e5. doi:10.1016/j.immuni.2018.11.003. PMC 7191997. PMID 30594464.
External links
- Description of the TGF beta producing genes at ncbi.nlm.nih.gov
- Diagram of the TGF beta signaling pathway at genome.ad.jp
- The TGF-beta system—Nature Reviews Molecular Cell Biology
- SMART:TGFB domain annotation—European Molecular Biology Laboratory Heidelberg
- TGF-beta at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
- Biochemists Solve Structure Of TGF-Beta And Its Receptor. 2008 – shows TGF-β3 dimer in TGFB-receptor
- Measurement of Human Latent TGF-β1
- TGF beta pathway diagram
- IntroPro Entry – IPR016319
2. The bone morphogenetic proteins and the growth differentiation factors
Bone morphogenetic proteins (BMPs) are a group of growth factors also known as cytokines and as metabologens. Originally discovered by their ability to induce the formation of bone and cartilage, BMPs are now considered to constitute a group of pivotal morphogenetic signals, orchestrating tissue architecture throughout the body. The important functioning of BMP signals in physiology is emphasized by the multitude of roles for dysregulated BMP signaling in pathological processes. Cancerous disease often involves misregulation of the BMP signaling system. Absence of BMP signaling is, for instance, an important factor in the progression of colon cancer, and conversely, overactivation of BMP signaling following reflux-induced esophagitis provokes Barrett’s esophagus and is thus instrumental in the development of esophageal adenocarcinoma.
- Reddi AH, Reddi A (2009). “Bone morphogenetic proteins (BMPs): from morphogens to metabologens”. Cytokine & Growth Factor Reviews. 20 (5–6): 341–2. doi:10.1016/j.cytogfr.2009.10.015. PMID 19900831.
- Bleuming SA, He XC, Kodach LL, Hardwick JC, Koopman FA, Ten Kate FJ, van Deventer SJ, Hommes DW, Peppelenbosch MP, Offerhaus GJ, Li L, van den Brink GR (Sep 2007). “Bone morphogenetic protein signaling suppresses tumorigenesis at gastric epithelial transition zones in mice”. Cancer Research. 67 (17): 8149–55. doi:10.1158/0008-5472.CAN-06-4659. PMID 17804727.
- Kodach LL, Wiercinska E, de Miranda NF, Bleuming SA, Musler AR, Peppelenbosch MP, Dekker E, van den Brink GR, van Noesel CJ, Morreau H, Hommes DW, Ten Dijke P, Offerhaus GJ, Hardwick JC (May 2008). “The bone morphogenetic protein pathway is inactivated in the majority of sporadic colorectal cancers”. Gastroenterology. 134 (5): 1332–41. doi:10.1053/j.gastro.2008.02.059. PMID 18471510.
- Milano F, van Baal JW, Buttar NS, Rygiel AM, de Kort F, DeMars CJ, Rosmolen WD, Bergman JJ, VAn Marle J, Wang KK, Peppelenbosch MP, Krishnadath KK (Jun 2007). “Bone morphogenetic protein 4 expressed in esophagitis induces a columnar phenotype in esophageal squamous cells”. Gastroenterology. 132 (7): 2412–21. doi:10.1053/j.gastro.2007.03.026. PMID 17570215.
BMPs interact with specific receptors on the cell surface, referred to as bone morphogenetic protein receptors (BMPRs).
Signal transduction through BMPRs results in mobilization of members of the SMAD family of proteins. The signaling pathways involving BMPs, BMPRs and SMADs are important in the development of the heart, central nervous system, and cartilage, as well as post-natal bone development.
They have an important role during embryonic development on the embryonic patterning and early skeletal formation. As such, disruption of BMP signaling can affect the body plan of the developing embryo. For example, BMP4 and its inhibitors noggin and chordin help regulate polarity of the embryo (i.e. back to front patterning). Specifically BMP-4 and its inhibitors play a major role in neurulation and the development of the neural plate. BMP-4 signals ectoderm cells to develop into skin cells, but the secretion of inhibitors by the underlying mesoderm blocks the action of BMP-4 to allow the ectoderm to continue on its normal course of neural cell development. Additionally, secretion of BMPs by the roof plate in the developing spinal cord helps to specify dorsal sensory interneurons.
- Andrews, Madeline (19 September 2017). “BMPs direct sensory interneuron identity in the developing spinal cord using signal- specific not morphogenic activities”. eLife. 6. doi:10.7554/eLife.30647. PMC 5605194. PMID 28925352.
As a member of the transforming growth factor-beta superfamily, BMP signaling regulates a variety of embryonic patterning during fetal and embryonic development. For example, BMP signaling controls the early formation of the Mullerian duct (MD) which is a tubular structure in early embryonic developmental stage and eventually becomes female reproductive tracts. Chemical inhibiting BMP signals in chicken embryo caused a disruption of MD invagination and blocked the epithelial thickening of the MD-forming region, indicating that the BMP signals play a role in early MD development. Moreover, BMP signaling is involved in the formation of foregut and hindgut, intestinal villus patterning, and endocardial differentiation. Villi contribute to increase the effective absorption of nutrients by extending the surface area in small intestine. Gain or lose function of BMP signaling altered the patterning of clusters and emergence of villi in mouse intestinal model. BMP signal derived from myocardium is also involved in endocardial differentiation during heart development. Inhibited BMP signal in zebrafish embryonic model caused strong reduction of endocardial differentiation, but only had little effect in myocardial development. In addition, Notch-Wnt-Bmp crosstalk is required for radial patterning during mouse cochlea development via antagonizing manner.
- Yuji, Yoshiko (2016). “Early formation of the Mullerian duct is regulated by sequential actions of BMP/Pax2 and FGF/Lim1 signaling”. Development. 143 (19): 3549–3559. doi:10.1242/dev.137067. PMID 27578782.
- Mariana, et al. (2017). “Genomic integration of Wnt/β-catenin and BMP/Smad1 signaling coordinates foregut and hindgut transcriptional programs”. Development. 144 (7): 1283–1295. doi:10.1242/dev.145789. PMC 5399627. PMID 28219948.
- Katherine, et al. (2016). “Villification in the mouse: Bmp signals control intestinal villus patterning”. Development. 143 (3): 427–436. doi:10.1242/dev.130112. PMC 4760312. PMID 26721501.
- Sharina, et al. (2015). “Myocardium and BMP signaling are required for endocardial differentiation”. Development. 142 (13): 2304–2315. doi:10.1242/dev.118687. PMC 4510589. PMID 26092845.
- Vidhya, et al. (2016). “Notch-Wnt-Bmp crosstalk regulates radial patterning in the mouse cochlea in a spatiotemporal manner”. Development. 143 (21): 4003–4015. doi:10.1242/dev.139469. PMC 5117145. PMID 27633988
Mutations in BMPs and their inhibitors are associated with a number of human disorders which affect the skeleton.
BMPs are also involved in adipogenesis and functional regulation of adipose tissue. BMP4 favors white adipogenesis, whereas BMP7 activates brown fat functionality; BMP inhibitors are also involved in this regulation.
- Blázquez-Medela, Ana M.; Jumabay, Medet; Boström, Kristina I. (2019-01-04). “Beyond the bone: Bone morphogenetic protein signaling in adipose tissue”. Obesity Reviews. 20 (5): 648–658. doi:10.1111/obr.12822. ISSN 1467-789X. PMC 6447448. PMID 30609449.
Types
Originally, seven such proteins were discovered. Of these, six (BMP2 through BMP7) belong to the Transforming growth factor beta superfamily of proteins. BMP1 is a metalloprotease. Since then, thirteen more BMPs, all of which are in the TGF-beta family, have been discovered, bringing the total to twenty.
- Even J, Eskander M, Kang J (Sep 2012). “Bone morphogenetic protein in spine surgery: current and future uses”. The Journal of the American Academy of Orthopaedic Surgeons. 20 (9): 547–52. doi:10.5435/JAAOS-20-09-547. PMID 22941797.
The current nomenclature only recognizes 13, as many others are put under the growth differentiation factor naming instead.
BMP | Known functions | Gene Locus |
---|---|---|
BMP1 | *BMP1 does not belong to the TGF-β family of proteins. It is a metalloprotease that acts on procollagen I, II, and III. It is involved in cartilage development. | Chromosome: 8; Location: 8p21 |
BMP2 | Acts as a disulfide-linked homodimer and induces bone and cartilage formation. It is a candidate as a retinoid mediator. Plays a key role in osteoblast differentiation. | Chromosome: 20; Location: 20p12 |
BMP3 | Induces bone formation. | Chromosome: 14; Location: 14p22 |
BMP4 | Regulates the formation of teeth, limbs and bone from mesoderm. It also plays a role in fracture repair, epidermis formation, dorsal-ventral axis formation, and ovarian follical development. | Chromosome: 14; Location: 14q22-q23 |
BMP5 | Performs functions in cartilage development. | Chromosome: 6; Location: 6p12.1 |
BMP6 | Plays a role in joint integrity in adults. Controls iron homeostasis via regulation of hepcidin. | Chromosome: 6; Location: 6p12.1 |
BMP7 | Plays a key role in osteoblast differentiation. It also induces the production of SMAD1. Also key in renal development and repair. | Chromosome: 20; Location: 20q13 |
BMP8a | Involved in bone and cartilage development. | Chromosome: 1; Location: 1p35–p32 |
BMP8b | Expressed in the hippocampus. | Chromosome: 1; Location: 1p35–p32 |
BMP10 | May play a role in the trabeculation of the embryonic heart. | Chromosome: 2; Location: 2p14 |
BMP11 | Controls anterior-posterior patterning. | Chromosome: 12; Location: 12p |
BMP15 | May play a role in oocyte and follicular development. | Chromosome: X; Location: Xp11.2 |
Several BMPs are also named ‘cartilage-derived morphogenetic proteins’ (CDMPs), while others are referred to as ‘growth differentiation factors‘ (GDFs).
Growth differentiation factors (GDFs) are a subfamily of proteins belonging to the transforming growth factor beta superfamily that have functions predominantly in development.
- Herpin A, Lelong C, Favrel P (2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Dev Comp Immunol. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
Several members of this subfamily have been described, and named GDF1 through GDF15.
- GDF1 is expressed chiefly in the nervous system and functions in left-right patterning and mesoderm induction during embryonic development.
- GDF2 (also known as BMP9) induces and maintains the response embryonic basal forebrain cholinergic neurons (BFCN) have to a neurotransmitter called acetylcholine, and regulates iron metabolism by increasing levels of a protein called hepcidin.
- Lopez-Coviella I, Follettie M, Mellott T, Kovacheva V, Slack B, Diesl V, Berse B, Thies R, Blusztajn J (2005). “Bone morphogenetic protein 9 induces the transcriptome of basal forebrain cholinergic neurons”. Proc Natl Acad Sci USA. 102 (19): 6984–9. Bibcode:2005PNAS..102.6984L. doi:10.1073/pnas.0502097102. PMC 1088172. PMID 15870197.
- Truksa J, Peng H, Lee P, Beutler E (2006). “Bone morphogenetic proteins 2, 4, and 9 stimulate murine hepcidin 1 expression independently of Hfe, transferrin receptor 2 (Tfr2), and IL-6”. Proc Natl Acad Sci USA. 103 (27): 10289–93. Bibcode:2006PNAS..10310289T. doi:10.1073/pnas.0603124103. PMC 1502450. PMID 16801541
- GDF3 is also known as “Vg-related gene 2” (Vgr-2). Expression of GDF3 occurs in ossifying bone during embryonic development and in the thymus, spleen, bone marrow brain, and adipose tissue of adults. It has a dual nature of function; it both inhibits and induces early stages of development in embryos.
- Levine A, Brivanlou A (2006). “GDF3 at the crossroads of TGF-beta signaling”. Cell Cycle. 5 (10): 1069–73. doi:10.4161/cc.5.10.2771. PMID 16721050.
- Levine A, Brivanlou A (2006). “GDF3, a BMP inhibitor, regulates cell fate in stem cells and early embryos”. Development. 133 (2): 209–16. doi:10.1242/dev.02192. PMID 16339188.
- Chen C, Ware S, Sato A, Houston-Hawkins D, Habas R, Matzuk M, Shen M, Brown C (2006). “The Vg1-related protein Gdf3 acts in a Nodal signaling pathway in the pre-gastrulation mouse embryo”. Development. 133 (2): 319–29. doi:10.1242/dev.02210. PMID 16368929.
- GDF5 is expressed in the developing central nervous system, with roles in the development of joints and the skeleton, and increasing the survival of neurones that respond to a neurotransmitter called dopamine.
- O’Keeffe G, Dockery P, Sullivan A (2004). “Effects of growth/differentiation factor 5 on the survival and morphology of embryonic rat midbrain dopaminergic neurones in vitro”. J Neurocytol. 33 (5): 479–88. doi:10.1007/s11068-004-0511-y. PMID 15906156. S2CID 25940876.
- Buxton P, Edwards C, Archer C, Francis-West P (2001). “Growth/differentiation factor-5 (GDF-5) and skeletal development”. J Bone Joint Surg Am. 83-A Suppl 1 (Pt 1): S23–30. PMID 11263662.
- Francis-West P, Parish J, Lee K, Archer C (1999). “BMP/GDF-signalling interactions during synovial joint development”. Cell Tissue Res. 296 (1): 111–9. doi:10.1007/s004410051272. PMID 10199971. S2CID 21942870.
- GDF6 interacts with bone morphogenetic proteins to regulate ectoderm patterning, and controls eye development.
- Chang C, Hemmati-Brivanlou A (1999). “Xenopus GDF6, a new antagonist of noggin and a partner of BMPs”. Development. 126 (15): 3347–57. doi:10.1242/dev.126.15.3347. PMID 10393114.
- Asai-Coakwell M, French C, Berry K, Ye M, Koss R, Somerville M, Mueller R, van Heyningen V, Waskiewicz A, Lehmann O (2007). “GDF6, a novel locus for a spectrum of ocular developmental anomalies”. American Journal of Human Genetics. 80 (2): 306–15. doi:10.1086/511280. PMC 1785352. PMID 17236135.
- Hanel M, Hensey C (2006). “Eye and neural defects associated with loss of GDF6”. BMC Dev Biol. 6: 43. doi:10.1186/1471-213X-6-43. PMC 1609107. PMID 17010201.
- GDF8 is now officially known as myostatin and controls the growth of muscle tissue.[14]
- GDF9, like GDF3, lacks one cysteine relative to other members of the TGF-β superfamily. Its gene expression is limited to the ovaries, and it has a role in ovulation.
- Juengel J, Bodensteiner K, Heath D, Hudson N, Moeller C, Smith P, Galloway S, Davis G, Sawyer H, McNatty K (2004). “Physiology of GDF9 and BMP15 signalling molecules”. Anim Reprod Sci. 82–83: 447–60. doi:10.1016/j.anireprosci.2004.04.021. PMID 15271472.
- Hreinsson J, Scott J, Rasmussen C, Swahn M, Hsueh A, Hovatta O (2002). “Growth differentiation factor-9 promotes the growth, development, and survival of human ovarian follicles in organ culture”. J Clin Endocrinol Metab. 87 (1): 316–21. doi:10.1210/jcem.87.1.8185. PMID 11788667
- GDF10 is closely related to BMP3 and has a roles in head formation and, it is presumed, in skeletal morphogenesis. It is also known as BMP-3b.
- Hino J, Kangawa K, Matsuo H, Nohno T, Nishimatsu S (2004). “Bone morphogenetic protein-3 family members and their biological functions”. Front Biosci. 9 (1–3): 1520–9. doi:10.2741/1355. PMID 14977563.
- Cunningham N, Jenkins N, Gilbert D, Copeland N, Reddi A, Lee S (1995). “Growth/differentiation factor-10: a new member of the transforming growth factor-beta superfamily related to bone morphogenetic protein-3”. Growth Factors. 12 (2): 99–109. doi:10.3109/08977199509028956. PMID 8679252.
- GDF11 controls anterior-posterior patterning by regulating the expression of Hox genes, and regulates the number of olfactory receptor neurons occurring in the olfactory epithelium, and numbers of retinal ganglionic cells developing in the retina.
- Andersson O, Reissmann E, Ibáñez C (2006). “Growth differentiation factor 11 signals through the transforming growth factor-beta receptor ALK5 to regionalize the anterior-posterior axis”. EMBO Reports. 7 (8): 831–7. doi:10.1038/sj.embor.7400752. PMC 1525155. PMID 16845371.
- Wu H, Ivkovic S, Murray R, Jaramillo S, Lyons K, Johnson J, Calof A (2003). “Autoregulation of neurogenesis by GDF11” (PDF). Neuron. 37 (2): 197–207. doi:10.1016/S0896-6273(02)01172-8. PMID 12546816. S2CID 15399794.
- Kim J, Wu H, Lander A, Lyons K, Matzuk M, Calof A (2005). “GDF11 controls the timing of progenitor cell competence in developing retina”. Science. 308 (5730): 1927–30. Bibcode:2005Sci…308.1927K. doi:10.1126/science.1110175. PMID 15976303. S2CID 42002862.
- GDF15 (also known as TGF-PL, MIC-1, PDF, PLAB, and PTGFB) has a role in regulating inflammatory and apoptotic pathways during tissue injury and certain disease processes.
- Zimmers T, Jin X, Hsiao E, McGrath S, Esquela A, Koniaris L (2005). “Growth differentiation factor-15/macrophage inhibitory cytokine-1 induction after kidney and lung injury”. Shock. 23 (6): 543–8. PMID 15897808.
- Hsiao E, Koniaris L, Zimmers-Koniaris T, Sebald S, Huynh T, Lee S (2000). “Characterization of growth-differentiation factor 15, a transforming growth factor beta superfamily member induced following liver injury”. Mol Cell Biol. 20 (10): 3742–51. doi:10.1128/MCB.20.10.3742-3751.2000. PMC 85678. PMID 10779363.
- Ago T, Sadoshima J (2006). “GDF15, a cardioprotective TGF-beta superfamily protein”. Circ Res. 98 (3): 294–7. doi:10.1161/01.RES.0000207919.83894.9d. PMID 16484622.
3. The activin and inhibin subfamilies
Activin and inhibin are two closely related protein complexes that have almost directly opposite biological effects. Identified in 1986, activin enhances FSH biosynthesis and secretion, and participates in the regulation of the menstrual cycle.
- Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, Karr D, Spiess J (1986). “Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid”. Nature. 321 (6072): 776–9. Bibcode:1986Natur.321..776V. doi:10.1038/321776a0. PMID 3012369. S2CID 4365045.
- Ling N, Ying SY, Ueno N, Shimasaki S, Esch F, Hotta M, Guillemin R (1986). “Pituitary FSH is released by a heterodimer of the beta-subunits from the two forms of inhibin”. Nature. 321 (6072): 779–82. Bibcode:1986Natur.321..779L. doi:10.1038/321779a0. PMID 3086749. S2CID 38100413.
Many other functions have been found to be exerted by activin, including roles in cell proliferation, differentiation, apoptosis, metabolism, homeostasis, immune response, wound repair, and endocrine function.
- Chen YG, Wang Q, Lin SL, Chang CD, Chuang J, Chung J, Ying SY (May 2006). “Activin signaling and its role in regulation of cell proliferation, apoptosis, and carcinogenesis”. Experimental Biology and Medicine. 231 (5): 534–44. doi:10.1177/153537020623100507. PMID 16636301. S2CID 39050907.
- Sulyok S, Wankell M, Alzheimer C, Werner S (October 2004). “Activin: an important regulator of wound repair, fibrosis, and neuroprotection”. Molecular and Cellular Endocrinology. 225 (1–2): 127–32. doi:10.1016/j.mce.2004.07.011. PMID 15451577. S2CID 6943949.
Conversely, inhibin downregulates FSH synthesis and inhibits FSH secretion.
- van Zonneveld P, Scheffer GJ, Broekmans FJ, Blankenstein MA, de Jong FH, Looman CW, Habbema JD, te Velde ER (March 2003). “Do cycle disturbances explain the age-related decline of female fertility? Cycle characteristics of women aged over 40 years compared with a reference population of young women”. Human Reproduction. 18 (3): 495–501. doi:10.1093/humrep/deg138. PMID 12615813.
The existence of inhibin was hypothesized as early as 1916; however, it was not demonstrated to exist until Neena Schwartz and Cornelia Channing‘s work in the mid-1970s, after which both proteins were molecularly characterized ten years later.
- Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WP, Schwartz NB, Mayo KE, Woodruff TK (October 2014). “Inhibin at 90: from discovery to clinical application, a historical review”. Endocrine Reviews. 35 (5): 747–94. doi:10.1210/er.2014-1003. PMC 4167436. PMID 25051334.
Activin is a dimer composed of two identical or very similar beta subunits. Inhibin is also a dimer wherein the first component is a beta subunit similar or identical to the beta subunit in activin. However, in contrast to activin, the second component of the inhibin dimer is a more distantly-related alpha subunit. Activin, inhibin and a number of other structurally related proteins such as anti-Müllerian hormone, bone morphogenetic protein, and growth differentiation factor belong to the TGF-β protein superfamily.
- Burger HG, Igarashi M (April 1988). “Inhibin: definition and nomenclature, including related substances”. The Journal of Clinical Endocrinology and Metabolism. 66 (4): 885–6. PMID 3346366.
- Robertson DM, Burger HG, Fuller PJ (March 2004). “Inhibin/activin and ovarian cancer”. Endocrine-Related Cancer. 11 (1): 35–49. doi:10.1677/erc.0.0110035. PMID 15027884. S2CID 12202820.
- Kingsley DM (January 1994). “The TGF-beta superfamily: new members, new receptors, and new genetic tests of function in different organisms”. Genes & Development. 8 (2): 133–46. doi:10.1101/gad.8.2.133. PMID 8299934.
Structure
The activin and inhibin protein complexes are both dimeric in structure, and, in each complex, the two monomers are linked to one another by a single disulfide bond.
- Ying SY (December 1987). “Inhibins and activins: chemical properties and biological activity”. Proceedings of the Society for Experimental Biology and Medicine. 186 (3): 253–64. doi:10.3181/00379727-186-42611a. PMID 3122219. S2CID 36872324.
In addition, both complexes are derived from the same family of related genes and proteins but differ in their subunit composition.
- Burger HG, Igarashi M (April 1988). “Inhibin: definition and nomenclature, including related substances”. The Journal of Clinical Endocrinology and Metabolism. 66 (4): 885–6. PMID 3346366.
The alpha and beta subunits share approximately 25% sequence similarity, whereas the similarity between beta subunits is approximately 65%.
- Kingsley DM (January 1994). “The TGF-beta superfamily: new members, new receptors, and new genetic tests of function in different organisms”. Genes & Development. 8 (2): 133–46. doi:10.1101/gad.8.2.133. PMID 8299934.
In mammals, four beta subunits have been described, called activin βA, activin βB, activin βC and activin βE. Activin βA and βB are identical to the two beta subunits of inhibin. A fifth subunit, activin βD, has been described in Xenopus laevis. Two activin βA subunits give rise to activin A, one βA, and one βB subunit gives rise to activin AB, and so on. Various, but not all theoretically possible, heterodimers have been described.
- Xu P, Hall AK (November 2006). “The role of activin in neuropeptide induction and pain sensation”. Developmental Biology. 299 (2): 303–9. doi:10.1016/j.ydbio.2006.08.026. PMID 16973148.
- Deli A, Kreidl E, Santifaller S, Trotter B, Seir K, Berger W, Schulte-Hermann R, Rodgarkia-Dara C, Grusch M (March 2008). “Activins and activin antagonists in hepatocellular carcinoma”. World Journal of Gastroenterology. 14 (11): 1699–709. doi:10.3748/wjg.14.1699. PMC 2695910. PMID 18350601.
The subunits are linked by a single covalent disulfide bond.
The βC subunit is able to form activin heterodimers with βA or βB subunits but is unable to dimerize with inhibin α.
- Mellor SL, Cranfield M, Ries R, Pedersen J, Cancilla B, de Kretser D, Groome NP, Mason AJ, Risbridger GP (December 2000). “Localization of activin beta(A)-, beta(B)-, and beta(C)-subunits in humanprostate and evidence for formation of new activin heterodimers of beta(C)-subunit”. The Journal of Clinical Endocrinology and Metabolism. 85 (12): 4851–8. doi:10.1210/jcem.85.12.7052. PMID 11134153.
Function
Activin
Activin is produced in the gonads, pituitary gland, placenta, and other organs:
- In the ovarian follicle, activin increases FSH binding and FSH-induced aromatization. It participates in androgen synthesis enhancing LH action in the ovary and testis. In the male, activin enhances spermatogenesis.
- Activin is strongly expressed in wounded skin, and overexpression of activin in epidermis of transgenicmice improves wound healing and enhances scar formation. Its action in wound repair and skin morphogenesis is through stimulation of keratinocytes and stromal cells in a dose-dependent manner.
- Bamberger, Casimir; Schärer, Agnes; Antsiferova, Maria; Tychsen, Birte; Pankow, Sandra; Müller, Mischa; Rülicke, Thomas; Paus, Ralf; Werner, Sabine (9 March 2021). “Activin Controls Skin Morphogenesis and Wound Repair Predominantly via Stromal Cells and in a Concentration-Dependent Manner via Keratinocytes”. The American Journal of Pathology. 167 (3): 733–747. doi:10.1016/S0002-9440(10)62047-0. PMC 1698729. PMID 16127153.
- Activin also regulates the morphogenesis of branching organs such as the prostate, lung, and especially kidney. Activin A increased the expression level of type-I collagen suggesting that activin A acts as a potent activator of fibroblasts.
- Lack of activin during development results in neural developmental defects.
- Upregulation of Activin A drives pluripotent stem cells into a mesoendodermal fate, and thus provides a useful tool for stem cell differentiation and organoid formation.
- Pauklin S, Vallier L (2015). “Activin/Nodal signalling in stem cells”. Development. 142 (4): 607–19. doi:10.1242/dev.091769. PMID 25670788.
Inhibin
In both females and males, inhibin inhibits FSH production. Inhibin does not inhibit the secretion of GnRH from the hypothalamus.
- Luisi S, Florio P, Reis FM, Petraglia F (2005). “Inhibins in female and male reproductive physiology: role in gametogenesis, conception, implantation and early pregnancy”. Human Reproduction Update. 11 (2): 123–35. doi:10.1093/humupd/dmh057. PMID 15618291.
- Le T, Bhushan V, Hofmann J (2012). First Aid for the USMLE Step 1. McGraw Hill. p. 534. ISBN 978-0-07-177636-3.
However, the overall mechanism differs between the sexes:
In females
Inhibin is produced in the gonads, pituitary gland, placenta, corpus luteum and other organs.
FSH stimulates the secretion of inhibin from the granulosa cells of the ovarian follicles in the ovaries. In turn, inhibin suppresses FSH.
- Inhibin B reaches a peak in the early- to mid-follicular phase, and a second peak at ovulation.
- Inhibin A reaches its peak in the mid-luteal phase.
Inhibin secretion is diminished by GnRH, and enhanced by insulin-like growth factor-1 (IGF-1).
In males
It is secreted from the Sertoli cells, located in the seminiferous tubules inside the testes.
- Skinner MK, McLachlan RI, Bremner WJ (October 1989). “Stimulation of Sertoli cell inhibin secretion by the testicular paracrine factor PModS”. Molecular and Cellular Endocrinology. 66 (2): 239–49. doi:10.1016/0303-7207(89)90036-1. hdl:1773/4395. PMID 2515083. S2CID 21885326.
Androgens stimulate inhibin production; this protein may also help to locally regulate spermatogenesis.
- Meachem SJ, Nieschlag E, Simoni M (November 2001). “Inhibin B in male reproduction: pathophysiology and clinical relevance”. European Journal of Endocrinology. 145 (5): 561–71. doi:10.1530/eje.0.1450561. PMID 11720872.
Mechanism of action
Activin
As with other members of the superfamily, activins interact with two types of cell surface transmembrane receptors (Types I and II) which have intrinsic serine/threonine kinase activities in their cytoplasmic domains:
Activin binds to the Type II receptor and initiates a cascade reaction that leads to the recruitment, phosphorylation, and activation of Type I activin receptor. This then interacts with and then phosphorylates SMAD2 and SMAD3, two of the cytoplasmic SMAD proteins.
Smad3 then translocates to the nucleus and interacts with SMAD4 through multimerization, resulting in their modulation as transcription factor complexes responsible for the expression of a large variety of genes.
Inhibin
In contrast to activin, much less is known about the mechanism of action of inhibin, but may involve competing with activin for binding to activin receptors and/or binding to inhibin-specific receptors.
- Robertson DM, Burger HG, Fuller PJ (March 2004). “Inhibin/activin and ovarian cancer”. Endocrine-Related Cancer. 11 (1): 35–49. doi:10.1677/erc.0.0110035. PMID 15027884. S2CID 12202820.
Clinical significance
Activin
Activin A is more plentiful in the adipose tissue of obese, compared to lean persons.
- Zaragosi LE, Wdziekonski B, Villageois P, Keophiphath M, Maumus M, Tchkonia T, Bourlier V, Mohsen-Kanson T, Ladoux A, Elabd C, Scheideler M, Trajanoski Z, Takashima Y, Amri EZ, Lacasa D, Sengenes C, Ailhaud G, Clément K, Bouloumie A, Kirkland JL, Dani C (2010). “Activin a plays a critical role in proliferation and differentiation of human adipose progenitors”. Diabetes. 59 (10): 2513–2521. doi:10.2337/db10-0013. PMC 3279533. PMID 20530742.
Activin A promotes the proliferation of adipocyte progenitor cells, while inhibiting their differentiation into adipocytes.
- Zaragosi LE, Wdziekonski B, Villageois P, Keophiphath M, Maumus M, Tchkonia T, Bourlier V, Mohsen-Kanson T, Ladoux A, Elabd C, Scheideler M, Trajanoski Z, Takashima Y, Amri EZ, Lacasa D, Sengenes C, Ailhaud G, Clément K, Bouloumie A, Kirkland JL, Dani C (2010). “Activin a plays a critical role in proliferation and differentiation of human adipose progenitors”. Diabetes. 59 (10): 2513–2521. doi:10.2337/db10-0013. PMC 3279533. PMID 20530742.
Activin A also increases inflammatory cytokines in macrophages.
- Zaragosi LE, Wdziekonski B, Villageois P, Keophiphath M, Maumus M, Tchkonia T, Bourlier V, Mohsen-Kanson T, Ladoux A, Elabd C, Scheideler M, Trajanoski Z, Takashima Y, Amri EZ, Lacasa D, Sengenes C, Ailhaud G, Clément K, Bouloumie A, Kirkland JL, Dani C (2010). “Activin a plays a critical role in proliferation and differentiation of human adipose progenitors”. Diabetes. 59 (10): 2513–2521. doi:10.2337/db10-0013. PMC 3279533. PMID 20530742.
A mutation in the gene for the activin receptor ACVR1 results in fibrodysplasia ossificans progressiva, a fatal disease that causes muscle and soft tissue to gradually be replaced by bone tissue. This condition is characterized by the formation of an extra skeleton that produces immobilization and eventually death by suffocation. The mutation in ACVR1 causes activin A, which normally acts as an antagonist of the receptor and blocks osteogenesis (bone growth), to behave as an agonist of the receptor and to induce hyperactive bone growth.
- Shore EM, Xu M, Feldman GJ, Fenstermacher DA, Cho TJ, Choi IH, Connor JM, Delai P, Glaser DL, LeMerrer M, Morhart R, Rogers JG, Smith R, Triffitt JT, Urtizberea JA, Zasloff M, Brown MA, Kaplan FS (May 2006). “A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva”. Nature Genetics. 38 (5): 525–527. doi:10.1038/ng1783. PMID 16642017. S2CID 41579747.
On 2 September 2015, Regeneron Pharmaceuticals announced that they had developed an antibody for activin A that effectively cures the disease in an animal model of the condition.
- Julie Steenhuysen (2 September 2015). “Regeneron scientists discover key to excess bone growth in rare disease”. Reuters.
Mutations in the ACVR1 gene have also been linked to cancer, especially diffuse intrinsic pontine glioma (DIPG).
- Taylor KR, Mackay A, Truffaux N, Butterfield YS, Morozova O, Philippe C, Castel D, Grasso CS, Vinci M, Carvalho D, Carcaboso AM, de Torres C, Cruz O, Mora J, Entz-Werle N, Ingram WJ, Monje M, Hargrave D, Bullock AN, Puget S, Yip S, Jones C, Grill J (May 2014). “Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma”. Nature Genetics. 46 (5): 457–61. doi:10.1038/ng.2925. PMC 4018681. PMID 24705252.
- “Cure Brain Cancer – News – Multiple Breakthroughs in Childhood Brain Cancer DIPG”. Cure Brain Cancer Foundation.
- Buczkowicz P, Hoeman C, Rakopoulos P, Pajovic S, Letourneau L, Dzamba M, et al. (May 2014). “Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations”. Nature Genetics. 46 (5): 451–6. doi:10.1038/ng.2936. PMC 3997489. PMID 24705254.
Elevated Activin B levels with normal Activin A levels provided a possible biomarker for myalgic encephalomyelitis/chronic fatigue syndrome.
- Lidbury BA, Kita B, Lewis DP, Hayward S, Ludlow H, Hedger MP, de Kretser DM (March 2017). “Activin B is a novel biomarker for chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) diagnosis: a cross sectional study”. Journal of Translational Medicine. 15 (1): 60. doi:10.1186/s12967-017-1161-4. PMC 5353946. PMID 28302133.
Activin A is overexpressed in many cancers. It was shown to promote tumorigenesis by hampering the adaptive anti-tumor immune response in melanoma.
- Donovan P, Dubey OA, Kallioinen S, Rogers KW, Muehlethaler K, Müller P, et al. (December 2017). “Paracrine Activin-A Signaling Promotes Melanoma Growth and Metastasis through Immune Evasion”. The Journal of Investigative Dermatology. 137 (12): 2578–2587. doi:10.1016/j.jid.2017.07.845. PMID 28844941.
Inhibin
Quantification of inhibin A is part of the prenatal quad screen that can be administered during pregnancy at a gestational age of 16–18 weeks. An elevated inhibin A (along with an increased beta-hCG, decreased AFP, and a decreased estriol) is suggestive of the presence of a fetus with Down syndrome.
- Aitken DA, Wallace EM, Crossley JA, Swanston IA, van Pareren Y, van Maarle M, Groome NP, Macri JN, Connor JM (May 1996). “Dimeric inhibin A as a marker for Down’s syndrome in early pregnancy”. The New England Journal of Medicine. 334 (19): 1231–6. doi:10.1056/NEJM199605093341904. PMID 8606718.
As a screening test, abnormal quad screen test results need to be followed up with more definitive tests.
It also has been used as a marker for ovarian cancer.
- Robertson DM, Pruysers E, Jobling T (April 2007). “Inhibin as a diagnostic marker for ovarian cancer”. Cancer Letters. 249 (1): 14–7. doi:10.1016/j.canlet.2006.12.017. PMID 17320281.
- Robertson DM, Pruysers E, Burger HG, Jobling T, McNeilage J, Healy D (October 2004). “Inhibins and ovarian cancer”. Molecular and Cellular Endocrinology. 225 (1–2): 65–71. doi:10.1016/j.mce.2004.02.014. PMID 15451569. S2CID 33801243
Inhibin B may be used as a marker of spermatogenesis function and male infertility. The mean serum inhibin B level is significantly higher among fertile men (approximately 140 pg/mL) than in infertile men (approximately 80 pg/mL).
- Myers GM, Lambert-Messerlian GM, Sigman M (December 2009). “Inhibin B reference data for fertile and infertile men in Northeast America”. Fertility and Sterility. 92 (6): 1920–3. doi:10.1016/j.fertnstert.2008.09.033. PMID 19006797.
In men with azoospermia, a positive test for inhibin B slightly raises the chances for successfully achieving pregnancy through testicular sperm extraction (TESE), although the association is not very substantial, having a sensitivity of 0.65 (95% confidence interval [CI]: 0.56–0.74) and a specificity of 0.83 (CI: 0.64–0.93) for prediction the presence of sperm in the testes in non-obstructive azoospermia.
- Toulis KA, Iliadou PK, Venetis CA, Tsametis C, Tarlatzis BC, Papadimas I, Goulis DG (2010). “Inhibin B and anti-Mullerian hormone as markers of persistent spermatogenesis in men with non-obstructive azoospermia: a meta-analysis of diagnostic accuracy studies”. Human Reproduction Update. 16 (6): 713–24. doi:10.1093/humupd/dmq024. PMID 20601364.
External links
- Activin at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
- Inhibin at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
- Grusch M, Kreidl E (1 August 2008). “Activin and follistatin in liver biology and hepatocellular carcinoma”. SciTopics. Elsevier. Archived from the original on 9 December 2008. Retrieved 24 December 2008.
4. The left-right determination factors
Lefty (left-right determination factors) are a class of proteins that are closely related members of the TGF-beta superfamily of growth factors. These proteins are secreted and play a role in left-right asymmetry determination of organ systems during development. Mutations of the genes encoding these proteins have been associated with left-right axis malformations, particularly in the heart and lungs.
- Hamada H, Meno C, Watanabe D, Saijoh Y (February 2002). “Establishment of vertebrate left-right asymmetry”. Nat. Rev. Genet. 3 (2): 103–13. doi:10.1038/nrg732. PMID 11836504. S2CID 20557143.
- Meno C, Shimono A, Saijoh Y, Yashiro K, Mochida K, Ohishi S, Noji S, Kondoh H, Hamada H (August 1998). “lefty-1 is required for left-right determination as a regulator of lefty-2 and nodal”. Cell. 94 (3): 287–97. doi:10.1016/S0092-8674(00)81472-5. PMID 9708731. S2CID 5666974.
Lefty, a divergent member of the transforming growth factor-β (TGF beta) superfamily of proteins, was originally discovered in the Hamada lab at the Osaka University using deletion screening of cDNA libraries in P19 embryonic carcinoma cells to find clones that did not differentiate when induced to differentiate using retinoic acid. From these screens, researchers found one gene that was a tentative member of the TGF-beta superfamily that was predominantly expressed on the left side the embryo and aptly named it lefty.
- Meno C, Saijoh Y, Fujii H, Ikeda M, Yokoyama T, Yokoyama M, Toyoda Y, Hamada H (May 1996). “Left-right asymmetric expression of the TGF beta-family member lefty in mouse embryos”. Nature. 381 (6578): 151–5. Bibcode:1996Natur.381..151M. doi:10.1038/381151a0. PMID 8610011. S2CID 4345275.
Like other members of the TGF-beta superfamily, lefty is synthesized as a preproprotein, meaning that the protein is proteolytically cleaved and excreted to produce the active form of the protein. However, lefty has only 20-25% sequence similarity with other members of the TGF-beta superfamily. Lefty is conserved in all vertebrates and many species have more than one homologue. Humans and mice, for instance have two homologues, Lefty 1 and Lefty 2, whose differential expression leads to distinct purposes while the mechanism of action is conserved.
- Kosaki K, Bassi MT, Kosaki R, Lewin M, Belmont J, Schauer G, Casey B (March 1999). “Characterization and mutation analysis of human LEFTY A and LEFTY B, homologues of murine genes implicated in left-right axis development”. Am. J. Hum. Genet. 64 (3): 712–21. doi:10.1086/302289. PMC 1377788. PMID 10053005
Lefty proteins function as an antagonist of the Nodal Signaling pathway. Nodal is another signaling protein which is responsible for gastrulation, left-right patterning and induction of the primitive node. As NODAL protein diffuse through an embryo, it triggers Nodal Signaling within tissues with the required receptors and coreceptors. Activated nodal signaling leads to the transcription of the lefty gene. The protein is then expressed, proteolytically cleaved, and finally secreted. Secreted lefty binds to EGF-CFC proteins like one-eyed pinhead in zebrafish keeping the essential cofactor from associating with NODAL/ Activin-like receptor complex. This will effectually block Nodal Signaling. During induction of the primitive streak, lefty confines Nodal activity to the posterior end of the embryo, establishing a posterior signaling center and inducing the formation of the primitive streak and mesoderm. (See Nodal Signaling or TGF beta signaling pathway for more information on the nodal signaling pathway.)
- Carlson, Bruce M. “Formation of Germ Layers and Early Derivatives.” Human Embryology and Developmental Biology. Philadelphia, Pennsylvania: Mosby/Elsevier, 2009. 91-95. Print.
- Schier AF (November 2009). “Nodal Morphogens”. Cold Spring Harb Perspect Biol. 1 (5): a003459. doi:10.1101/cshperspect.a003459. PMC 2773646. PMID 20066122.
There are many differences between the left and right sides, including heart and lung positioning. Mutations in these genes cause incorrect positioning of these organs (e.g., situs inversus), or in the case of constitutively inactive lefty, the embryo becomes entirely mesoderm and fails to pattern or develop. During vertebrate development, lefty proteins regulate left-right asymmetry by controlling the spatiotemporal influence of the NODAL protein. Lefty1 in the ventral midline prevents the Cerberus (paracrine factor or “Caronte”) signal from passing to the right side of the embryo.
- Hamada H, Meno C, Watanabe D, Saijoh Y (February 2002). “Establishment of vertebrate left-right asymmetry”. Nat. Rev. Genet. 3 (2): 103–13. doi:10.1038/nrg732. PMID 11836504. S2CID 20557143
This spatiotemporal control is achieved by using two sources of excreted lefty. While lefty is produced in response to activated nodal signaling, it is also produced and secreted in the anterior visceral endoderm (AVE). The balance of lefty from the AVE and from Nodal Signaling results in the patterning of the embryo and left-right asymmetry.
- Takaoka K, Yamamoto M, Hamada H (August 2007). “Origin of body axes in the mouse embryo”. Curr. Opin. Genet. Dev. 17 (4): 344–50. doi:10.1016/j.gde.2007.06.001. PMID 17646095.
Proper functioning of Lefty is crucial to the proper development of the heart, lungs, spleen, and liver. Mutations in Lefty, called Lefty-A, are associated with left-right patterning defects. This mutation may cause congenital heart defects due to malformation, interrupted inferior vena cava, and lack of lung asymmetry (left pulmonary isomerism).
- Carlson, Bruce M. “Formation of Germ Layers and Early Derivatives.” Human Embryology and Developmental Biology. Philadelphia, Pennsylvania: Mosby/Elsevier, 2009. 91-95. Print.
Lefty2 may play a role in endometrial bleeding.
- Kothapalli R, Buyuksal I, Wu SQ, Chegini N, Tabibzadeh S (May 1997). “Detection of ebaf, a novel human gene of the transforming growth factor beta superfamily association of gene expression with endometrial bleeding”. J. Clin. Invest. 99 (10): 2342–50. doi:10.1172/JCI119415. PMC 508072. PMID 9153275.
- Tabibzadeh S (2005). “Role of EBAF/Lefty in implantation and uterine bleeding”. Ernst Schering Res. Found. Workshop. Ernst Schering Research Foundation Workshop. 52 (52): 159–89. doi:10.1007/3-540-27147-3_8. ISBN 978-3-540-23089-2. PMID 15704472.
Lefty-1 is a regulatory gene that plays a vital role in the determination of the left-right internal asymmetry observed in mammals. The lefty-1 protein works in tandem with two other genes: lefty-2 and nodal. As the primitive node migrates towards the cranial end of the embryo during development, its cilia preferentially sling lefty-2 and nodal towards the left side of the embryo.
- Hashimoto M, Shinohara K, Wang J, Ikeuchi S, Yoshiba S, Meno C, Nonaka S, Takada S, Hatta K, Wynshaw-Boris A, Hamada H (February 2010). “Planar polarization of node cells determines the rotational axis of node cilia”. Nature Cell Biology. 12 (2): 170–6. doi:10.1038/ncb2020. PMID 20098415. S2CID 6379844.
These two genes encode for “leftness”, and initiate the formation of the heart, spleen, and other internal organs that are found on the left side in a typical human being. Lefty-1 protein can be viewed as a barrier between the left and right portions of the embryo that prevents the diffusion of lefty-2 and nodal to the right side. This ensures that the left-determining molecules are confined to their correct developmental domain. A variety of defects were observed in mice that had lefty-1 deleted, including left pulmonary isomerism, situs inversus, and atrial septal defect.
- Meno C, Shimono A, Saijoh Y, Yashiro K, Mochida K, Ohishi S, Noji S, Kondoh H, Hamada H (August 1998). “lefty-1 is required for left-right determination as a regulator of lefty-2 and nodal”. Cell. 94 (3): 287–97. doi:10.1016/S0092-8674(00)81472-5. PMID 9708731. S2CID 5666974.
The high incidence of left pulmonary isomerism in the knockout mice indicates that lefty-1 itself is not involved in encoding for leftness, but simply ensures the correct compartmentation of the left-determining molecules. In the absence of the lefty-1 barrier, lefty-2 and nodal are free to diffuse to the right side and initiate the development of a left lung that was meant to be limited to the left side of the thoracic cavity.
Further reading
- Carlson BM (2014). “Formation of germ layers and early derivatives.”. Human Embryology and Developmental Biology. Philadelphia, Pennsylvania: Mosby/Elsevier. pp. 75–91. ISBN 978-0-323-08279-2.
- Sakuma R, Ohnishi Yi Y, Meno C, Fujii H, Juan H, Takeuchi J, Ogura T, Li E, Miyazono K, Hamada H (April 2002). “Inhibition of Nodal signalling by Lefty mediated through interaction with common receptors and efficient diffusion”. Genes to Cells: Devoted to Molecular & Cellular Mechanisms. 7 (4): 401–12. doi:10.1046/j.1365-2443.2002.00528.x. PMID 11952836. S2CID 19320756.
4. A group encompassing various divergent members?
Transforming growth factor-beta (TGF-beta) is a multifunctional peptide that controls proliferation, differentiation and other functions in many cell types.
- Roberts AB, Sporn MB (1990). Peptide growth factors and their receptors. Berlin: Springer-Verlag. ISBN 3-540-51184-9.
TGF-beta-1 is a peptide of 112 amino acid residues derived by proteolytic cleavage from the C-terminal of a precursor protein. These proteins interact with a conserved family of cell surface serine/threonine-specific protein kinase receptors, and generate intracellular signals using a conserved family of proteins called SMADs. They play fundamental roles in the regulation of basic biological processes such as growth, development, tissue homeostasis and regulation of the immune system.
- Herpin A, Lelong C, Favrel P (May 2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Dev. Comp. Immunol. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
Structure
Proteins from the TGF-beta superfamily are only active as homo- or heterodimer; the two chains being linked by a single disulfide bond. From X-ray studies of TGF-beta-2, it is known that all the other cysteines are involved in intrachain disulfide bonds.
- Daopin S, Piez KA, Ogawa Y, Davies DR (July 1992). “Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily”. Science. 257 (5068): 369–73. Bibcode:1992Sci…257..369D. doi:10.1126/science.1631557. PMID 1631557.
As shown in the following schematic representation, there are four disulfide bonds in the TGF-beta’s and in inhibin beta chains, while the other members of this superfamily lack the first bond.
interchain | +------------------------------------------|+ | || xxxxcxxxxxCcxxxxxxxxxxxxxxxxxxCxxCxxxxxxxxxxxxxxxxxxxCCxxxxxxxxxxxxxxxxxxxCxCx | | | | | | +------+ +--|----------------------------------------+ | +------------------------------------------+
where ‘C’ denotes a conserved cysteine involved in a disulfide bond.
Examples
Human genes encoding proteins that contain this domain include:
AMH; ARTN; BMP2; BMP3; BMP4; BMP5; BMP6; BMP7; BMP8A; BMP8B; BMP10; BMP15; GDF1; GDF2; GDF3; GDF5; GDF6; GDF7; GDF9; GDF10; GDF11; GDF15; GDNF; INHA; INHBA; INHBB; INHBC; INHBE; LEFTY1; LEFTY2; MSTN; NODAL; NRTN; PSPN; TGFB1; TGFB2; TGFB3;
References
- Schlunegger MP, Grütter MG (July 1992). “An unusual feature revealed by the crystal structure at 2.2 A resolution of human transforming growth factor-beta 2”. Nature. 358 (6385): 430–4. Bibcode:1992Natur.358..430S. doi:10.1038/358430a0. PMID 1641027. S2CID 4239431.
- Assoian RK, Komoriya A, Meyers CA, Miller DM, Sporn MB (June 1983). “Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization”. J. Biol. Chem. 258 (11): 7155–60. doi:10.1016/S0021-9258(18)32345-7. PMID 6602130.
- Herpin A, Lelong C, Favrel P (May 2004). “Transforming growth factor-beta-related proteins: an ancestral and widespread superfamily of cytokines in metazoans”. Dev. Comp. Immunol. 28 (5): 461–85. doi:10.1016/j.dci.2003.09.007. PMID 15062644.
- Burt DW (April 1992). “Evolutionary grouping of the transforming growth factor-beta superfamily”. Biochem. Biophys. Res. Commun. 184 (2): 590–5. doi:10.1016/0006-291X(92)90630-4. PMID 1575734.
- Burt DW, Law AS (1994). “Evolution of the transforming growth factor-beta superfamily” (PDF). Prog. Growth Factor Res. 5 (1): 99–118. doi:10.1016/0955-2235(94)90020-5. hdl:20.500.11820/50fc2d69-c411-4835-9cd9-36bf4144bae4. PMID 8199356. S2CID 41326578.
- Roberts AB, Sporn MB (1990). Peptide growth factors and their receptors. Berlin: Springer-Verlag. ISBN 3-540-51184-9.
- Daopin S, Piez KA, Ogawa Y, Davies DR (July 1992). “Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily”. Science. 257 (5068): 369–73. Bibcode:1992Sci…257..369D. doi:10.1126/science.1631557. PMID 1631557.
- “Prosite Documentation PDOC00223”. Archived from the original on 2011-05-25. Retrieved 2006-07-01.
- Fliesler SJ, Kisselev OG (26 December 2007). Signal Transduction in the Retina. CRC Press. pp. 273–. ISBN 978-1-4200-0716-9.
- Thiriet M (14 December 2011). Signaling at the Cell Surface in the Circulatory and Ventilatory Systems. Springer Science & Business Media. pp. 666–. ISBN 978-1-4614-1991-4.
- Wrana JL, Attisano L, Cárcamo J, et al. (December 1992). “TGF beta signals through a heteromeric protein kinase receptor complex”. Cell. 71 (6): 1003–14. doi:10.1016/0092-8674(92)90395-S. PMID 1333888. S2CID 54397586.
- Huse, M; Muir, TW; Xu, L; Chen, YG; Kuriyan, J; Massagué, J (September 2001). “The TGF beta receptor activation process: an inhibitor- to substrate-binding switch”. Molecular Cell. 8 (3): 671–82. doi:10.1016/S1097-2765(01)00332-X. PMID 11583628.
- Andres JL, Stanley K, et al. (1989). “Membrane-anchored and soluble forms of betaglycan, a polymorphic proteoglycan that binds transforming growth factor-beta”. J. Cell Biol. 109 (6 (Pt 1)): 3137–3145. doi:10.1083/jcb.109.6.3137. PMC 2115961. PMID 2592419.
- Andres JL, DeFalcis D, et al. (1992). “Binding of two growth factor families to separate domains of the proteoglycan betaglycan”. J. Biol. Chem. 267 (9): 5927–5930. PMID 1556106.
Intercellular signaling peptides and proteins / ligands |
---|
Kinases: Serine/threonine-specific protein kinases (EC 2.7.11-12) |
---|
Drugs for treatment of bone diseases (M05) |
---|
- Developmental genes and proteins
- TGFβ domain
- Protein domains
- Membrane proteins
- Protein superfamilies
- Genes on human chromosome 1
- Genes on human chromosome 2
- Genes on human chromosome 7
- Genes on human chromosome 12
- Genes on human chromosome 14
- Genes on human chromosome 19
- Mammal reproductive system
- Hormones of the ovary
- Peptide hormones
- Human female endocrine system
- Bone morphogenetic protein
- Growth factors
- Animal developmental biology
- Osaka University research
Leave a Reply