GDF11

From WikiProjectMed
Jump to navigation Jump to search
GDF11
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesGDF11, BMP-11, BMP11, growth differentiation factor 11, VHO
External IDsOMIM: 603936 MGI: 1338027 HomoloGene: 21183 GeneCards: GDF11
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_005811

NM_010272

RefSeq (protein)

NP_005802

NP_034402

Location (UCSC)Chr 12: 55.74 – 55.76 MbChr 10: 128.72 – 128.73 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

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.[5] GDF11 is a member of the Transforming growth factor beta family.[6]

GDF11 acts as a cytokine and its sequence is highly conserved between in humans, mice and rats.[7] 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.[8]

Tissue distribution

GDF11 is expressed in many tissues, including skeletal muscle, pancreas, kidney, nervous system, and retina.[6]

Function

Gene deletion and over-expression studies indicate that GDF11 primarily regulates the embryological development of the skeletal system. It may also help regulate development of the central nervous system, blood vessels, the kidney and other tissues.[9][10][11][12][13]

GDF11 improves neurodegenerative and neurovascular disease outcomes, increases skeletal muscle volume, and enhances muscle strength. Its wide-ranging biological effects may include the reversal of senescence in clinical applications, as well as the ability to reverse age-related pathological changes and regulate organ regeneration after injury.[14]

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.[15] It determines Hox gene expression domains and rostrocaudal identity in the caudal spinal cord.[12]

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.[16] This cytokine also inhibits the proliferation of olfactory receptor neural progenitors to regulate the number of neurons in the olfactory epithelium,[17] and controls the competence of progenitor cells to regulate numbers of retinal ganglionic cells developing in the retina.[18] Other studies in mice suggest that GDF11 is involved in mesodermal formation and neurogenesis during embryonic development.

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.[15] It is also closely related to myostatin, a negative regulator of muscle growth,[19][20] both structurally and phylogenetically.[21]

Though GDF11 is 90% structurally similar to myostatin, GDF11's mechanism of action is opposite that of myostatin since it declines with age and exerts anti-aging regenerative effects in skeletal muscle in mice[22]

Human studies

GDF11 levels fall to zero in humans at a mean age of 73.71. No endogenous GDF11 production results in the cessation of stem cell DNA repair which causes stem cells to die off and their populations fall to zero at an even faster rate. Since one cannot survive without hematopoietic, mesenchymal, etc. stems cells, this suggests that GDF11 may play a key role in maximum lifespan determination.[23]

Elevian, whose founders include Harvard Stem Cell Institute researchers Dr. Amy Wagers, Dr. Lee Rubin and Dr. Rich Lee, has raised $58 million in two rounds of funding to study GDF11. On June 19, 2022, the New York Times published an article about GDF11 and Elevian titled "Can a 'Magic' Protein Slow the Aging Process?". The article stated that Elevian will conduct clinical trials using GDF11 to repair stroke damage in humans starting in Q1 of 2023.[24]

Physical fitness correlated with GDF11 levels in serum, which is in line with results of a previous study reporting higher serum GDF11 in lifelong exercising men compared to their lifelong sedentary peers. Physical fitness not only determines the tissue-specific expression and concentration of GDF11, but also the magnitude of its exercise-stimulated regulation.[25]

GDF11 levels in individuals with major depressive disorder are significantly lower compared to healthy controls. Administration of GDF11 in aged mice stimulates neuronal autophagy which improves memory and alleviates senescence and depression-like symptoms in a neurogenesis-independent manner.[26]

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.[27]

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.

Note that GDF11 levels can increase in response to various cellular stressors, including hypoxia (low oxygen levels) and inflammation. Tumor microenvironments often have low oxygen levels and increased inflammation, which could be the cause higher GDF11 expression in colon cancer patients..[27]

Animal studies

In 2014, GDF11 was described as a life extension factor in two publications based on the results of parabiosis experiments in mice [28][29] that were chosen as Science's scientific breakthrough of the year.[30] Later studies questioned these findings.[31][32][33][34] Researchers disagree on the selectivity of the tests used to measure GDF11 and on the activity of GDF11 from various commercially available sources.[35] 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.

GDF11 is a powerful senolytic and antioxidant. GDF11 fed mice saw 45.7% reduction in senescent liver cells and a 21.7% reduction in senescent kidney cells. GDF11 induces generation of antioxidant enzymes (CAT, SOD and GPX), which directly results in reduction of ROS levels, which then decelerates protein oxidation, lipid peroxidation and possibly LF and SA-β-Gal development, which in turn extends lifespan of aged mice.[36]

GDF11 attenuates the senescence of ovarian and testicular cells, and contributes to the recovery of ovarian and testicular endocrine functions. Moreover, GDF11 could rescue the diminished ovarian reserve in female mice and enhance the activities of marker enzymes of testicular function (SDH and G6PD) in male mice, suggesting a potential improvement of fertility.[37]

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.[38]

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.[39]

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.[40]

GDF11 contributes to limiting functional damage of mitochondria in cardiomyocytes (heart cells) following ischemic (lack of blood flow) injury or anoxia (oxygen deprivation) insult, and repressing apoptosis in mitochondria-dependent and mitochondria-independent manners by increasing telomerase activities. This suggests that GDF11 may be an effective treatment for post heart attack patients.[41]

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.[42]

GDF11 improves endothelial dysfunction, decreases endothelial apoptosis, and reduces inflammation, consequently decreases atherosclerotic plaques area in apolipoprotein E−/− mice.[43]

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.[44]

GDF11 improves tubular regeneration after acute kidney injury in elderly mice. Supplementing GDF11 increased tubular cell dedifferentiation and proliferation as well as improved the prognosis of old mice that underwent ischemia–reperfusion injury by upregulating the ERK1/2 signaling pathway.[45]

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.[46]

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.[47]

This GDF11 paper summarizes GDF11 expression in various organs as well as a table showing effects of GDF11 in cardiac, muscle skeletal and nervous system disease.[48]

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.[28]

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.[49]

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,[50]

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.[51]

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).[27]

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000135414Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000025352Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ 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): 5846–5858. doi:10.1128/MCB.25.14.5846-5858.2005. PMC 1168807. PMID 15988002.
  6. ^ a b 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.
  7. ^ 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.
  8. ^ "Gene GDF11". Genecards. Retrieved 25 May 2013.
  9. ^ Egerman MA, Glass DJ (April 2019). "The role of GDF11 in aging and skeletal muscle, cardiac and bone homeostasis". Critical Reviews in Biochemistry and Molecular Biology. 54 (2): 174–183. doi:10.1080/10409238.2019.1610722. PMID 31144559. S2CID 169039791.
  10. ^ Esquela AF, Lee SJ (May 2003). "Regulation of metanephric kidney development by growth/differentiation factor 11". Developmental Biology. 257 (2): 356–370. doi:10.1016/s0012-1606(03)00100-3. PMID 12729564.
  11. ^ Dichmann DS, Yassin H, Serup P (November 2006). "Analysis of pancreatic endocrine development in GDF11-deficient mice". Developmental Dynamics. 235 (11): 3016–3025. doi:10.1002/dvdy.20953. PMID 16964608. S2CID 30675774.
  12. ^ a b Liu JP (August 2006). "The function of growth/differentiation factor 11 (Gdf11) in rostrocaudal patterning of the developing spinal cord". Development. 133 (15): 2865–2874. doi:10.1242/dev.02478. PMID 16790475.
  13. ^ 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): 407–420. doi:10.1006/dbio.2000.9981. PMID 11203700.
  14. ^ Ma Y, Liu Y, Han F, Qiu H, Shi J, Huang N, et al. (April 2021). "Growth differentiation factor 11: a "rejuvenation factor" involved in regulation of age-related diseases?". Aging. 13 (8): 12258–12272. doi:10.18632/aging.202881. PMC 8109099. PMID 33886503. S2CID 233372437.
  15. ^ a b 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): 831–837. doi:10.1038/sj.embor.7400752. PMC 1525155. PMID 16845371.
  16. ^ 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): 260–264. doi:10.1038/10320. PMID 10391213. S2CID 1172738.
  17. ^ Wu HH, Ivkovic S, Murray RC, Jaramillo S, Lyons KM, Johnson JE, et al. (January 2003). "Autoregulation of neurogenesis by GDF11". Neuron. 37 (2): 197–207. doi:10.1016/S0896-6273(02)01172-8. PMID 12546816. S2CID 15399794.
  18. ^ 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): 1927–1930. Bibcode:2005Sci...308.1927K. doi:10.1126/science.1110175. PMID 15976303. S2CID 42002862.
  19. ^ 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): 12457–12461. Bibcode:1997PNAS...9412457M. doi:10.1073/pnas.94.23.12457. PMC 24998. PMID 9356471.
  20. ^ Lee SJ, McPherron AC (October 1999). "Myostatin and the control of skeletal muscle mass". Current Opinion in Genetics & Development. 9 (5): 604–607. doi:10.1016/S0959-437X(99)00004-0. PMID 10508689.
  21. ^ Kerr T, Roalson EH, Rodgers BD (2005). "Phylogenetic analysis of the myostatin gene sub-family and the differential expression of a novel member in zebrafish". Evolution & Development. 7 (5): 390–400. doi:10.1111/j.1525-142X.2005.05044.x. PMID 16174033. S2CID 6538603.
  22. ^ Añón-Hidalgo J, Catalán V, Rodríguez A, Ramírez B, Silva C, Galofré JC, et al. (March 2019). "Circulating GDF11 levels are decreased with age but are unchanged with obesity and type 2 diabetes". Aging. 11 (6): 1733–1744. doi:10.18632/aging.101865. PMC 6461177. PMID 30897065.
  23. ^ Delgado D, Bilbao AM, Beitia M, Garate A, Sánchez P, González-Burguera I, et al. (February 2021) [11/24/202]. "Effects of Platelet-Rich Plasma on Cellular Populations of the Central Nervous System: The Influence of Donor Age". International Journal of Molecular Sciences. 22 (4): 1725. doi:10.3390/ijms22041725. PMC 7915891. PMID 33572157.
  24. ^ Zimmerman E (2022-07-19). "Can a 'Magic' Protein Slow the Aging Process?". The New York Times. ISSN 0362-4331. Retrieved 2022-12-05.
  25. ^ Schön M, Marček Malenovská K, Nemec M, Alchus Laiferová N, Straka I, Košutzká Z, et al. (2023). "Acute endurance exercise modulates growth differentiation factor 11 in cerebrospinal fluid of healthy young adults". Frontiers in Endocrinology. 14. doi:10.3389/fendo.2023.1137048. ISSN 1664-2392. PMC 10073538. PMID 37033257.
  26. ^ Moigneu C, Abdellaoui S, Ramos-Brossier M, Pfaffenseller B, Wollenhaupt-Aguiar B, de Azevedo Cardoso T, et al. (2023-02-02). "Systemic GDF11 attenuates depression-like phenotype in aged mice via stimulation of neuronal autophagy". Nature Aging. 3 (2): 213–228. doi:10.1038/s43587-022-00352-3. ISSN 2662-8465. PMC 10154197. PMID 37118117.
  27. ^ a b c Simoni-Nieves A, Gerardo-Ramírez M, Pedraza-Vázquez G, Chávez-Rodríguez L, Bucio L, Souza V, et al. (2019-10-15). "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.
  28. ^ a b 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): 649–652. Bibcode:2014Sci...344..649S. doi:10.1126/science.1251152. PMC 4104429. PMID 24797481.
  29. ^ 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): 630–634. Bibcode:2014Sci...344..630K. doi:10.1126/science.1251141. PMC 4123747. PMID 24797482.
  30. ^ "'Young blood' reverses aging – the breakthrough of 2014 #GDF11". 2015-01-05.
  31. ^ 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): 164–174. doi:10.1016/j.cmet.2015.05.010. PMC 4497834. PMID 26001423.
  32. ^ Age-reversal effects of 'young blood' molecule GDF-11 called into question, retrieved 20 May 2015
  33. ^ Reardon S (2015), "'Young blood' anti-ageing mechanism called into question", Nature, doi:10.1038/nature.2015.17583, S2CID 182418356, retrieved 20 May 2015
  34. ^ 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): 926–932. doi:10.1161/CIRCRESAHA.115.307527. PMC 4636963. PMID 26383970.
  35. ^ Kaiser J (Oct 2015). "Antiaging protein is the real deal, Harvard team claims". Science. doi:10.1126/science.aad4748.
  36. ^ Song L, Wu F, Li C, Zhang S (June 2022). "Dietary intake of GDF11 delays the onset of several biomarkers of aging in male mice through anti-oxidant system via Smad2/3 pathway". Biogerontology. 23 (3): 341–362. doi:10.1007/s10522-022-09967-w. PMC 9125541. PMID 35604508.
  37. ^ Zhou Y, Ni S, Li C, Song L, Zhang S (May 2022). "Gonadal Rejuvenation of Mice by Growth Differentiation Factor 11". The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 77 (5): 892–901. doi:10.1093/gerona/glab343. PMID 34791251.
  38. ^ 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): 6163–6174. doi:10.1242/dev.01535. PMID 15548585.
  39. ^ Katsimpardi L, Kuperwasser N, Camus C, Moigneu C, Chiche A, Tolle V, et al. (January 2020). "Systemic GDF11 stimulates the secretion of adiponectin and induces a calorie restriction-like phenotype in aged mice". Aging Cell. 19 (1): e13038. doi:10.1111/acel.13038. PMC 6974718. PMID 31637864.
  40. ^ 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.
  41. ^ Chen L, Luo G, Liu Y, Lin H, Zheng C, Xie D, et al. (2021-07-02). "Growth differentiation factor 11 attenuates cardiac ischemia reperfusion injury via enhancing mitochondrial biogenesis and telomerase activity". Cell Death & Disease. 12 (7): 665. doi:10.1038/s41419-021-03954-8. ISSN 2041-4889. PMC 8253774. PMID 34215721.
  42. ^ Zhao Y, Zhu J, Zhang N, Liu Q, Wang Y, Hu X, et al. (October 2020). "GDF11 enhances therapeutic efficacy of mesenchymal stem cells for myocardial infarction via YME1L-mediated OPA1 processing". Stem Cells Translational Medicine. 9 (10): 1257–1271. doi:10.1002/sctm.20-0005. PMC 7519765. PMID 32515551.
  43. ^ Mei W, Xiang G, Li Y, Li H, Xiang L, Lu J, et al. (November 2016). "GDF11 Protects against Endothelial Injury and Reduces Atherosclerotic Lesion Formation in Apolipoprotein E-Null Mice". Molecular Therapy. 24 (11): 1926–1938. doi:10.1038/mt.2016.160. PMC 5154476. PMID 27502608.
  44. ^ Dai Z, Song G, Balakrishnan A, Yang T, Yuan Q, Möbus S, et al. (June 2020). "Growth differentiation factor 11 attenuates liver fibrosis via expansion of liver progenitor cells". Gut. 69 (6): 1104–1115. doi:10.1136/gutjnl-2019-318812. PMC 7282557. PMID 31767630.
  45. ^ Zhang Y, Li Q, Liu D, Huang Q, Cai G, Cui S, et al. (October 2016). "GDF11 improves tubular regeneration after acute kidney injury in elderly mice". Scientific Reports. 6 (1): 34624. Bibcode:2016NatSR...634624Z. doi:10.1038/srep34624. PMC 5050408. PMID 27703192.
  46. ^ 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.
  47. ^ Rochette L, Mazini L, Meloux A, Zeller M, Cottin Y, Vergely C, et al. (April 2020). "Anti-Aging Effects of GDF11 on Skin". International Journal of Molecular Sciences. 21 (7): 2598. doi:10.3390/ijms21072598. PMC 7177281. PMID 32283613.
  48. ^ Ma Y, Liu Y, Han F, Qiu H, Shi J, Huang N, et al. (April 2021). "Growth differentiation factor 11: a "rejuvenation factor" involved in regulation of age-related diseases?". Aging. 13 (8): 12258–12272. doi:10.18632/aging.202881. PMC 8109099. PMID 33886503.
  49. ^ 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): 828–839. doi:10.1016/j.cell.2013.04.015. PMC 3677132. PMID 23663781.
  50. ^ 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. S2CID 59607006.
  51. ^ Liu Y, Shao L, Chen K, Wang Z, Wang J, Jing W, et al. (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.

Further reading

External links