GDF11

Growth differentiation factor 11 (GDF11) also known as bone morphogenetic protein 11 (BMP-11) is a protein that in humans is encoded by the growth differentiation factor 11 gene.[4] GDF11 is a member of the Transforming growth factor beta family.[5]

GDF11
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesGDF11, BMP-11, BMP11, growth differentiation factor 11
External IDsOMIM: 603936 MGI: 1338027 HomoloGene: 21183 GeneCards: GDF11
Gene location (Human)
Chr.Chromosome 12 (human)[1]
Band12q13.2Start55,743,122 bp[1]
End55,757,264 bp[1]
Orthologs
SpeciesHumanMouse
Entrez

10220

14561

Ensembl

ENSG00000135414

n/a

UniProt

O95390

Q9Z1W4

RefSeq (mRNA)

NM_005811

NM_010272

RefSeq (protein)

NP_005802

NP_034402

Location (UCSC)Chr 12: 55.74 – 55.76 Mbn/a
PubMed search[2][3]
Wikidata
View/Edit HumanView/Edit Mouse

GDF11 acts as a cytokine and its molecular structure is identical in humans, mice and rats.[6] The bone morphogenetic protein group is characterized by a polybasic proteolytic processing site, which is cleaved to produce a protein containing seven conserved cysteine residues.[7]

Tissue distribution

GDF11 is expressed in many tissues, including skeletal muscle, pancreas, kidney, nervous system, and retina.[5]

Function

GDF11 expression is elevated in young animals. GDF11 has pro-neurogenic, pro-angiogenic properties, induces CNS patterning and has been proposed as a "rejuvenating factor"[8][9]

Animal studies

Systemic GDF11 treatment improves vasculature in the hippocampus and cortex of old mice resulting in enhanced neurogenesis.[10] Also, systematic replenishment of GDF11 improved the survival and morphology of β-cells and improved glucose metabolism in both non genetic and genetic mouse models of type 2 diabetes.[11]

GDF11 enhances therapeutic efficacy of mesenchymal stem cells for myocardial Infarction. This novel role of GDF11 may be used for a new approach of stem cell therapy for myocardial infarction. [12]

GDF11 triggers a calorie restriction‐like phenotype without affecting appetite or GDF15 levels in the blood, restores the insulin/IGF‐1 signaling pathway, and stimulates adiponectin secretion from white adipose tissue by direct action on adipocytes, while repairing neurogenesis in the aged brain.[13]

GDF11 gene transfer alleviates HFD-induced obesity, hyperglycemia, insulin resistance, and fatty liver development. In obese and STZ-induced diabetic mice, GDF11 gene transfer restores glucose metabolism and improves insulin resistance.[14]

GDF11 attenuates liver fibrosis via expansion of liver progenitor cells. The protective role of GDF11 during liver fibrosis and suggest a potential application of GDF11 for the treatment of chronic liver disease.[15]

GDF11 is a regulator of skin biology and has significant effects on the production of procollagen I and hyaluronic acid. GDF11 also activates the Smad2/3 phosphorylation pathway in skin endothelial cells and improves skin vasculature.[16]

GDF11 exerts considerable anti-aging effects on skin. As the key member of the TGF-Beta superfamily, GDF11 represents a promising therapeutic agent for the treatment of a number of inflammatory skin diseases, including psoriasis.[17]

Supplementation of systemic GDF11 levels, which normally decline with age, by heterochronic parabiosis or systemic delivery of recombinant protein, reversed functional impairments and restored genomic integrity in aged muscle stem cells (satellite cells). Increased GDF11 levels in aged mice also improved muscle structural and functional features and increased strength and endurance exercise capacity.[18]

Treatment of old mice to restore GDF11 to youthful levels recapitulated the effects of parabiosis and reversed age-related hypertrophy, revealing a therapeutic opportunity for cardiac aging.[19]

GDF11 has been found to reduce oxidative stress and was able to reduce the levels of AGEs, protein oxidation and lipid peroxidation, and to slow down the accumulation of age-related histological markers. GDF11 significantly prevented the decrease in CAT, GPX and SOD activities,[20]

Therapeutic application of GDF11 mounts a protective response against liver fibrosis by increasing the number of LGR5+ progenitor cells in the liver. The protective role of GDF11 during liver fibrosis and suggest a potential application of GDF11 for the treatment of chronic liver disease.[21]

Enhanced GDF11 expression promoted apoptosis and down-regulated GDF11 expression inhibited apoptosis in pancreatic cancer cell lines. These findings suggested that GDF11 acted as a tumor suppressor for pancreatic cancer.[22]

GDF11 induces tumor suppressive properties in human hepatocellular carcinoma-derived cells, Huh7 and Hep3B cell lines, restricting spheroid formation and clonogenic capacity, an effect that is also observed in other liver cancer cell lines (SNU-182, Hepa1-6, and HepG2), decreasing proliferation, motogenesis, and invasion. Similarly, Bajikar et al. (23) identified a tumor-suppressive role of GDF11 in a triple-negative breast cancer (TNBC).[23]

It has been reported that GDF11 is down-regulated in pancreatic cancer tissue, compared with surrounding tissue, and pancreatic cell lines exhibit a low expression of the growth factor (65). This group also reported that, in a cohort of 63 PC patients, those with high GDF11 expression had significantly better survival rates in comparison with those with low GDF11 expression. These effects were related to decreased proliferation, migration and invasion, and these observations are in agreement with those reported in HCC and TNBC. GDF11 is also capable of inducing apoptosis in pancreatic cancer cell lines.[23]

However, In 130 patients with colorectal cancer (CRC), the expression of GDF11 was significantly higher compared with normal tissue (56). The classification of the patient cohort in low and high GDF11 expression revealed that those patients with high levels of GDF11 showed a higher frequency of lymph node metastasis, more deaths and lower survival. The study suggests that GDF11 could be a prognostic biomarker in patients with this disease[24]

In 2014, GDF11 was described as a life extension factor in two publications based on the results of a parabiosis experiments with mice [18][25] that were chosen as Science's scientific breakthrough of the year.[26] Later studies questioned these findings.[27][28][29][30] Researchers disagree on the selectivity of the tests used to measure GDF11 and on the activity of GDF11 from various commercially available sources.[31] The full relationship of GDF11 to aging—and any possible differences in the action of GDF11 in mice, rats, and humans—is unclear and continues to be researched.

Effects on cell growth and differentiation

GDF11 belongs to the transforming growth factor beta superfamily that controls anterior-posterior patterning by regulating the expression of Hox genes.[32] It determines Hox gene expression domains and rostrocaudal identity in the caudal spinal cord.[33]

During mouse development, GDF11 expression begins in the tail bud and caudal neural plate region. GDF knock-out mice display skeletal defects as a result of patterning problems with anterior-posterior positioning.[34]

In the mouse adult central nervous system, GDF11 alone can improve the cerebral vasculature and enhance neurogenesis.[25]

This cytokine also inhibits the proliferation of olfactory receptor neuron progenitors to regulate the number of olfactory receptor neurons occurring in the olfactory epithelium,[35] and controls the competence of progenitor cells to regulate numbers of retinal ganglionic cells developing in the retina.[36]

Other studies in mice suggest that GDF11 is involved in mesodermal formation and neurogenesis during embryonic development. The members of this TGF-β superfamily are involved in the regulation of cell growth and differentiation not only in embryonic tissues, but adult tissues as well.[37]

GDF11 can bind type I TGF-beta superfamily receptors ACVR1B (ALK4), TGFBR1 (ALK5) and ACVR1C (ALK7), but predominantly uses ALK4 and ALK5 for signal transduction.[32]

GDF11 is closely related to myostatin, a negative regulator of muscle growth.[38][39] Both myostatin and GDF11 are involved in the regulation of cardiomyocyte proliferation.

GDF11 is a regulator of kidney organogenesis,[40] pancreatic development,[41] the rostro-caudal patterning in the development of spinal cords,[33] and of chondrogenesis.[42]

Due to the similarities between myostatin and GDF11, the actions of GDF11 are likely regulated by WFIKKN2, a large extracellular multidomain protein consisting of follistatin, immunoglobulin, protease inhibitor, and NTR domains.[43] WFIKKN2 has a high affinity for GDF11, and previously has been found to inhibit the biological activities of myostatin.[44]

Effect on cardiac and skeletal muscle aging

GDF11 has been identified as a blood circulating factor that has the ability to reverse age-related cardiac hypertrophy in mice. GDF11 gene expression and protein abundance decreases with age, and it shows differential abundance between young and old mice in parabiosis procedures, causing youthful regeneration of cardiomyocytes, a reduction in the brain natriuretic peptide (BNP) and in the atrial natriuretic peptide (ANP). GDF11 also causes an increase in expression of SERCA-2, an enzyme necessary for relaxation during diastolic functions.[19] GDF11 activates the TGF-β pathway in cardiomyocytes derived from pluripotent hematopoietic stem cells and suppresses the phosphorylation of Forkhead (FOX proteins) transcription factors. These effects suggest an "anti-hypertrophic effect", aiding in the reversal process of age-related hypertrophy, on the cardiomyocytes.[19] In 2014, peripheral supplementation of GDF11 protein (in mice) was shown to ameliorate the age-related dysfunction of skeletal muscle by rescuing the function of aged muscle stem cells. In humans, older males who had been chronically active over their lives show higher concentrations of GDF11 than inactive older men, and the concentration of circulating GDF11 correlated with leg power output when cycling.[45] These results have led to claims that GDF11 may be an anti-aging rejuvenation factor.[18]

These previous findings have been disputed since another publication has demonstrated the contrary, concluding that GDF11 increases with age and has deleterious effects on skeletal muscle regeneration,[27] being a pro-aging factor, with very high levels in some aged individuals. However, in October 2015, a Harvard study showed these contrary results to be the result of a flawed assay that was detecting immunoglobulin and not GDF11. The Harvard study claimed GDF11 does in fact reverse age-related cardiac hypertrophy.[31] However the Harvard study both ignored the GDF11-specific assay that was developed, establishing that GDF11 in mice is undetectable, and that the factor measured was in fact myostatin.[27] Also, the Harvard study combined the measure of GDF11 and GDF8 (myostatin), using a non-specific antibody, further confusing matters.

In 2016 conflicting reviews from different research teams were published concerning the effects of GDF11 on skeletal and cardiac muscle.[46] [47] One of the reviews reported an anti-hypertrophic effect in aging mice,[46] but the other team denied that cardiac hypertrophy occurs in old mice, asserting that GDF11 causes muscle wasting.[47] Both teams agreed that whether GDF11 increases or decreases with age had not been established.[46][47] A 2017 study found that super-physiological levels of GDF11 induced muscle wasting in the skeletal muscle of mice.[48]

References

  1. GRCh38: Ensembl release 89: ENSG00000135414 - Ensembl, May 2017
  2. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  3. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. Ge G, Hopkins DR, Ho WB, Greenspan DS (July 2005). "GDF11 forms a bone morphogenetic protein 1-activated latent complex that can modulate nerve growth factor-induced differentiation of PC12 cells". Molecular and Cellular Biology. 25 (14): 5846–58. doi:10.1128/MCB.25.14.5846-5858.2005. PMC 1168807. PMID 15988002.
  5. Simoni-Nieves A, Gerardo-Ramírez M, Pedraza-Vázquez G, Chávez-Rodríguez L, Bucio L, Souza V, Miranda-Labra RU, Gomez-Quiroz LE, Gutiérrez-Ruiz MC (2019). "GDF11 Implications in Cancer Biology and Metabolism. Facts and Controversies". Frontiers in Oncology. 9: 1039. doi:10.3389/fonc.2019.01039. PMC 6803553. PMID 31681577.
  6. Jamaiyar A, Wan W, Janota DM, Enrick MK, Chilian WM, Yin L (July 2017). "The versatility and paradox of GDF 11". Pharmacology & Therapeutics. 175: 28–34. doi:10.1016/j.pharmthera.2017.02.032. PMC 6319258. PMID 28223232.
  7. "Gene GDF11". Genecards. Retrieved 25 May 2013.
  8. Rochette L, Malka G (July 2019). "Neuroprotective Potential of GDF11: Myth or Reality?". International Journal of Molecular Sciences. 20 (14): 3563. doi:10.3390/ijms20143563. PMC 6679312. PMID 31330871.
  9. Schafer MJ, LeBrasseur NK (February 2019). "The influence of GDF11 on brain fate and function". GeroScience. 41 (1): 1–11. doi:10.1007/s11357-019-00054-6. PMC 6423340. PMID 30729414.
  10. Ozek C, Krolewski RC, Buchanan SM, Rubin LL (November 2018). "Growth Differentiation Factor 11 treatment leads to neuronal and vascular improvements in the hippocampus of aged mice". Scientific Reports. 8 (1): 17293. Bibcode:2018NatSR...817293O. doi:10.1038/s41598-018-35716-6. PMC 6251885. PMID 30470794.
  11. Harmon EB, Apelqvist AA, Smart NG, Gu X, Osborne DH, Kim SK (December 2004). "GDF11 modulates NGN3+ islet progenitor cell number and promotes beta-cell differentiation in pancreas development". Development. 131 (24): 6163–74. doi:10.1242/dev.01535. PMID 15548585.
  12. Zhao Y, Zhu J, Zhang N, Liu Q, Wang Y, Hu X, et al. (June 2020). "GDF11 enhances therapeutic efficacy of mesenchymal stem cells for myocardial infarction via YME1L-mediated OPA1 processing". Stem Cells Translational Medicine. doi:10.1002/sctm.20-0005. PMID 32515551.
  13. Katsimpardi L, Kuperwasser N, Camus C, Moigneu C, Chiche A, Tolle V, et al. (2020). "Systemic GDF11 stimulates the secretion of adiponectin and induces a calorie restriction-like phenotype in aged mice". Aging Cell. 0 (1): e13038. doi:10.1111/acel.13038. PMC 6974718. PMID 31637864.
  14. Lu B, Zhong J, Pan J, Yuan X, Ren M, Jiang L, et al. (December 2019). "Gdf11 gene transfer prevents high fat diet-induced obesity and improves metabolic homeostasis in obese and STZ-induced diabetic mice". Journal of Translational Medicine. 17 (1): 422. doi:10.1186/s12967-019-02166-1. PMC 6915940. PMID 31847906.
  15. Dai, Zhen; Song, Guangqi; Balakrishnan, Asha; Yang, Taihua; Yuan, Qinggong; Möbus, Selina; Weiss, Anna-Carina; Bentler, Martin; Zhu, Jimin; Jiang, Xuemei; Shen, Xizhong (2020-06-01). "Growth differentiation factor 11 attenuates liver fibrosis via expansion of liver progenitor cells". Gut. 69 (6): 1104–1115. doi:10.1136/gutjnl-2019-318812. ISSN 0017-5749. PMID 31767630.
  16. Idkowiak-Baldys J, Santhanam U, Buchanan SM, Pfaff KL, Rubin LL, Lyga J (2019-06-10). "Growth differentiation factor 11 (GDF11) has pronounced effects on skin biology". PLOS ONE. 14 (6): e0218035. Bibcode:2019PLoSO..1418035I. doi:10.1371/journal.pone.0218035. PMC 6557520. PMID 31181098.
  17. Rochette, Luc; Mazini, Loubna; Meloux, Alexandre; Zeller, Marianne; Cottin, Yves; Vergely, Catherine; Malka, Gabriel (January 2020). "Anti-Aging Effects of GDF11 on Skin". International Journal of Molecular Sciences. 21 (7): 2598. doi:10.3390/ijms21072598. PMC 7177281. PMID 32283613.
  18. Sinha M, Jang YC, Oh J, Khong D, Wu EY, Manohar R, et al. (May 2014). "Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle". Science. 344 (6184): 649–52. Bibcode:2014Sci...344..649S. doi:10.1126/science.1251152. PMC 4104429. PMID 24797481.
  19. Loffredo FS, Steinhauser ML, Jay SM, Gannon J, Pancoast JR, Yalamanchi P, et al. (May 2013). "Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy". Cell. 153 (4): 828–39. doi:10.1016/j.cell.2013.04.015. PMC 3677132. PMID 23663781.
  20. Zhou Y, Song L, Ni S, Zhang Y, Zhang S (August 2019). "Administration of rGDF11 retards the aging process in male mice via action of anti-oxidant system". Biogerontology. 20 (4): 433–443. doi:10.1007/s10522-019-09799-1. PMID 30726519.
  21. Dai Z, Song G, Balakrishnan A, Yang T, Yuan Q, Möbus S, et al. (November 2019). "Growth differentiation factor 11 attenuates liver fibrosis via expansion of liver progenitor cells". Gut. 69 (6): gutjnl-2019-318812. doi:10.1136/gutjnl-2019-318812. PMID 31767630.
  22. Liu Y, Shao L, Chen K, Wang Z, Wang J, Jing W, Hu M (2018-11-27). "GDF11 restrains tumor growth by promoting apoptosis in pancreatic cancer". OncoTargets and Therapy. 11: 8371–8379. doi:10.2147/OTT.S181792. PMC 6267626. PMID 30568460.
  23. Simoni-Nieves, Arturo; Gerardo-Ramírez, Monserrat; Pedraza-Vázquez, Gibrán; Chávez-Rodríguez, Lisette; Bucio, Leticia; Souza, Verónica; Miranda-Labra, Roxana U.; Gomez-Quiroz, Luis E.; Gutiérrez-Ruiz, María Concepción (2019-10-15). "GDF11 Implications in Cancer Biology and Metabolism. Facts and Controversies". Frontiers in Oncology. 9. doi:10.3389/fonc.2019.01039. ISSN 2234-943X. PMC 6803553. PMID 31681577.
  24. Simoni-Nieves A, Gerardo-Ramírez M, Pedraza-Vázquez G, Chávez-Rodríguez L, Bucio L, Souza V, et al. (2019). "GDF11 Implications in Cancer Biology and Metabolism. Facts and Controversies". Frontiers in Oncology. 9: 1039. doi:10.3389/fonc.2019.01039. PMC 6803553. PMID 31681577.
  25. Katsimpardi L, Litterman NK, Schein PA, Miller CM, Loffredo FS, Wojtkiewicz GR, et al. (May 2014). "Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors". Science. 344 (6184): 630–4. Bibcode:2014Sci...344..630K. doi:10.1126/science.1251141. PMC 4123747. PMID 24797482.
  26. "'Young blood' reverses aging – the breakthrough of 2014 #GDF11". 2015-01-05.
  27. Egerman MA, Cadena SM, Gilbert JA, Meyer A, Nelson HN, Swalley SE, et al. (July 2015). "GDF11 Increases with Age and Inhibits Skeletal Muscle Regeneration". Cell Metabolism. 22 (1): 164–74. doi:10.1016/j.cmet.2015.05.010. PMC 4497834. PMID 26001423.
  28. Age-reversal effects of 'young blood' molecule GDF-11 called into question, retrieved 20 May 2015
  29. Reardon, Sara (2015), "'Young blood' anti-ageing mechanism called into question", Nature, doi:10.1038/nature.2015.17583, retrieved 20 May 2015
  30. Smith SC, Zhang X, Zhang X, Gross P, Starosta T, Mohsin S, et al. (November 2015). "GDF11 does not rescue aging-related pathological hypertrophy". Circulation Research. 117 (11): 926–32. doi:10.1161/CIRCRESAHA.115.307527. PMC 4636963. PMID 26383970.
  31. Kaiser J (Oct 2015). "Antiaging protein is the real deal, Harvard team claims". Science. doi:10.1126/science.aad4748.
  32. Andersson O, Reissmann E, Ibáñez CF (August 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.
  33. Liu JP (August 2006). "The function of growth/differentiation factor 11 (Gdf11) in rostrocaudal patterning of the developing spinal cord". Development. 133 (15): 2865–74. doi:10.1242/dev.02478. PMID 16790475.
  34. McPherron AC, Lawler AM, Lee SJ (July 1999). "Regulation of anterior/posterior patterning of the axial skeleton by growth/differentiation factor 11". Nature Genetics. 22 (3): 260–4. doi:10.1038/10320. PMID 10391213.
  35. Wu HH, Ivkovic S, Murray RC, Jaramillo S, Lyons KM, Johnson JE, Calof AL (January 2003). "Autoregulation of neurogenesis by GDF11" (PDF). Neuron. 37 (2): 197–207. doi:10.1016/S0896-6273(02)01172-8. PMID 12546816.
  36. Kim J, Wu HH, Lander AD, Lyons KM, Matzuk MM, Calof AL (June 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.
  37. "GDF11". Genecards.
  38. McPherron AC, Lee SJ (November 1997). "Double muscling in cattle due to mutations in the myostatin gene". Proceedings of the National Academy of Sciences of the United States of America. 94 (23): 12457–61. Bibcode:1997PNAS...9412457M. doi:10.1073/pnas.94.23.12457. PMC 24998. PMID 9356471.
  39. Lee SJ, McPherron AC (October 1999). "Myostatin and the control of skeletal muscle mass". Current Opinion in Genetics & Development. 9 (5): 604–7. doi:10.1016/S0959-437X(99)00004-0. PMID 10508689.
  40. Esquela AF, Lee SJ (May 2003). "Regulation of metanephric kidney development by growth/differentiation factor 11". Developmental Biology. 257 (2): 356–70. doi:10.1016/s0012-1606(03)00100-3. PMID 12729564.
  41. Dichmann DS, Yassin H, Serup P (November 2006). "Analysis of pancreatic endocrine development in GDF11-deficient mice". Developmental Dynamics. 235 (11): 3016–25. doi:10.1002/dvdy.20953. PMID 16964608.
  42. Gamer LW, Cox KA, Small C, Rosen V (January 2001). "Gdf11 is a negative regulator of chondrogenesis and myogenesis in the developing chick limb". Developmental Biology. 229 (2): 407–20. doi:10.1006/dbio.2000.9981. PMID 11203700.
  43. Kondás K, Szláma G, Trexler M, Patthy L (August 2008). "Both WFIKKN1 and WFIKKN2 have high affinity for growth and differentiation factors 8 and 11". The Journal of Biological Chemistry. 283 (35): 23677–84. doi:10.1074/jbc.M803025200. PMC 3259755. PMID 18596030.
  44. "WJIKKN2". Geneards. Retrieved 25 May 2013.
  45. Elliott BT, Herbert P, Sculthorpe N, Grace FM, Stratton D, Hayes LD (July 2017). "Lifelong exercise, but not short-term high-intensity interval training, increases GDF11, a marker of successful aging: a preliminary investigation". Physiological Reports. 5 (13): e13343. doi:10.14814/phy2.13343. PMC 5506528. PMID 28701523.
  46. Walker RG, Poggioli T, Katsimpardi L, Buchanan SM, Oh J, Wattrus S, et al. (April 2016). "Biochemistry and Biology of GDF11 and Myostatin: Similarities, Differences, and Questions for Future Investigation". Circulation Research. 118 (7): 1125–41, discussion 1142. doi:10.1161/CIRCRESAHA.116.308391. PMC 4818972. PMID 27034275.
  47. Harper SC, Brack A, MacDonnell S, Franti M, Olwin BB, Bailey BA, et al. (April 2016). "Is Growth Differentiation Factor 11 a Realistic Therapeutic for Aging-Dependent Muscle Defects?". Circulation Research. 118 (7): 1143–50, discussion 1150. doi:10.1161/CIRCRESAHA.116.307962. PMC 4829942. PMID 27034276.
  48. Hammers DW, Merscham-Banda M, Hsiao JY, Engst S, Hartman JJ, Sweeney HL (April 2017). "Supraphysiological levels of GDF11 induce striated muscle atrophy". EMBO Molecular Medicine. 9 (4): 531–544. doi:10.15252/emmm.201607231. PMC 5376753. PMID 28270449.

Further reading

This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.