quadraScope

Prevention and treatment of age-related diseases


MENU

Oncology

Relevant papers published by Drs. Carmela Abraham, Cidi Chen, Ella Zeldich and collaborators include:

View the Article
Aging-suppressor Klotho: Prospects in diagnostics and therapeutics

Papers by others

  1. Doi, S., et al., Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem, 2011. 286(10): p. 8655- 8665.
  2. Yee, D., Targeting insulin-like growth factor pathways. Br J Cancer, 2006. 94(4): p. 465-8.
  3. Wolf, I., et al., Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene, 2008. 27(56): p. 7094-105.
  4. Li, X.X., et al., Klotho suppresses growth and invasion of colon cancer cells through inhibition of IGF1R-mediated PI3K/AKT pathway. Int J Oncol, 2014. 45(2): p. 611-8.
  5. Abramovitz, L., et al., KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res, 2011. 17(13): p. 4254-66.
  6. Chen, B., et al., Inhibition of lung cancer cells growth, motility and induction of apoptosis by Klotho, a novel secreted Wnt antagonist, in a dose-dependent manner. Cancer Biol Ther, 2012. 13(12): p. 1221-8.
  7. Jiang, B., Y. Gu, and Y. Chen, Identification of novel predictive markers for the prognosis of pancreatic ductal adenocarcinoma. Cancer Invest, 2014. 32(6): p. 218-25.
  8. Arbel Rubinstein, T., et al., Klotho suppresses colorectal cancer through modulation of the unfolded protein response. Oncogene, 2019. 38(6): p. 794-807.
  9. Lojkin, I., et al., Reduced expression and growth inhibitory activity of the aging suppressor klotho in epithelial ovarian cancer. Cancer Lett, 2015. 362(2): p. 149-57.
  10. Chen, B., et al., Overexpression of Klotho Inhibits HELF Fibroblasts SASP-related Protumoral Effects on Non-small Cell Lung Cancer Cells. J Cancer, 2018. 9(7): p. 1248- 1258.
  11. Chen, B., et al., Klotho inhibits growth and promotes apoptosis in human lung cancer cell line A549. J Exp Clin Cancer Res, 2010. 29(1): p. 99.
  12. Wang, X., et al., Combined effects of klotho and soluble CD40 ligand on A549 lung cancer cells. Oncol Rep, 2011. 25(5): p. 1465-72.
  13. Rubinek, T. and I. Wolf, The Role of Alpha-Klotho as a Universal Tumor Suppressor. Vitam Horm, 2016. 101: p. 197-214.
  14. Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation. Cell, 2011. 144(5): p. 646-74.
  15. Shmulevich, R., et al., Klotho rewires cellular metabolism of breast cancer cells through alteration of calcium shuttling and mitochondrial activity. Oncogene, 2020. 39(24): p. 4636- 4649.
  16. Delcroix, V., et al., The Role of the Anti-Aging Protein Klotho in IGF-1 Signaling and Reticular Calcium Leak: Impact on the Chemosensitivity of Dedifferentiated Liposarcomas. Cancers (Basel), 2018. 10(11).
  17. Ligumsky, H., et al., Tumor Suppressor Activity of Klotho in Breast Cancer Is Revealed by Structure-Function Analysis. Mol Cancer Res, 2015. 13(10): p. 1398-407.
  18. Pan, J., et al., Klotho, an anti-senescence related gene, is frequently inactivated through promoter hypermethylation in colorectal cancer. Tumour Biol, 2011. 32(4): p. 729-35.
  19. Tang, X., et al., Klotho: a tumor suppressor and modulator of the Wnt/β-catenin pathway in human hepatocellular carcinoma. Lab Invest, 2016. 96(2): p. 197-205.
  20. Shu, G., et al., Restoration of klotho expression induces apoptosis and autophagy in hepatocellular carcinoma cells. Cell Oncol (Dordr), 2013. 36(2): p. 121-9.
  21. Sun, H., et al., Overexpression of Klotho suppresses liver cancer progression and induces cell apoptosis by negatively regulating wnt/β-catenin signaling pathway. World J Surg Oncol, 2015. 13: p. 307.
  22. Wang, L., et al., Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res, 2011. 1(1): p. 111-119.
  23. Xie, B., et al., Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int, 2013. 13(1): p. 18.
  24. Chang, B., et al., Klotho inhibits the capacity of cell migration and invasion in cervical cancer. Oncol Rep, 2012. 28(3): p. 1022-8.
  25. Xie, B., et al., Hmgb1 inhibits Klotho expression and malignant phenotype in melanoma cells by activating NF-κB. Oncotarget, 2016. 7(49): p. 80765-80782.
  26. Zhou, X., et al., Klotho, an anti-aging gene, acts as a tumor suppressor and inhibitor of IGF- 1R signaling in diffuse large B cell lymphoma. J Hematol Oncol, 2017. 10(1): p. 37.
  27. Lee, J., et al., The anti-aging gene KLOTHO is a novel target for epigenetic silencing in human cervical carcinoma. Mol Cancer, 2010. 9: p. 109.
  28. Camilli, T.C., et al., Loss of Klotho during melanoma progression leads to increased filamin cleavage, increased Wnt5A expression, and enhanced melanoma cell motility. Pigment Cell Melanoma Res, 2011. 24(1): p. 175-86.
  29. Yan, Y., et al., Reduced Klotho expression contributes to poor survival rates in human patients with ovarian cancer, and overexpression of Klotho inhibits the progression of ovarian cancer partly via the inhibition of systemic inflammation in nude mice. Mol Med Rep, 2017. 15(4): p. 1777-1785.
  30. Arbel Rubinstein, T., et al., A Transgenic Model Reveals the Role of Klotho in Pancreatic Cancer Development and Paves the Way for New Klotho-Based Therapy. Cancers (Basel), 2021. 13(24).
  31. Rubinek, T., et al., Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat, 2012. 133(2): p. 649-57.
  32. Gigante, M., et al., Soluble Serum αKlotho Is a Potential Predictive Marker of Disease Progression in Clear Cell Renal Cell Carcinoma. Medicine (Baltimore), 2015. 94(45): p. e1917.
  33. Tang, X., et al., Expression of klotho and β-catenin in esophageal squamous cell carcinoma, and their clinicopathological and prognostic significance. Dis Esophagus, 2016. 29(3): p. 207-14.
  34. Wu, Q., et al., Klotho Inhibits Proliferation in a RET Fusion Model of Papillary Thyroid Cancer by Regulating the Wnt/β-Catenin Pathway. Cancer Manag Res, 2021. 13: p. 4791- 4802.
  35. Lu, L., et al., Klotho expression in epithelial ovarian cancer and its association with insulin- like growth factors and disease progression. Cancer Invest, 2008. 26(2): p. 185-92.
  36. Zhu, Y., et al., Klotho suppresses tumor progression via inhibiting PI3K/Akt/GSK3β/Snail signaling in renal cell carcinoma. Cancer Sci, 2013. 104(6): p. 663-71.
  37. Pawlikowski, M., et al., Expression of α-Klotho protein in human thyroid cancers - an immunohistochemical study. Endokrynol Pol, 2019. 70(3): p. 237-240.
  38. Suvannasankha, A., et al., FGF23 is elevated in multiple myeloma and increases heparanase expression by tumor cells. Oncotarget, 2015. 6(23): p. 19647-60.
  39. Usuda, J., et al., Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer, 2011. 74(2): p. 332-7.
  40. Usuda, J., et al., Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer, 2011. 72(3): p. 355-9.
  41. Peshes-Yeloz, N., et al., Role of Klotho Protein in Tumor Genesis, Cancer Progression, and Prognosis in Patients with High-Grade Glioma. World Neurosurg, 2019. 130: p. e324- e332.
  42. Zhu, Y., et al., DNA methylation-mediated Klotho silencing is an independent prognostic biomarker of head and neck squamous carcinoma. Cancer Manag Res, 2019. 11: p. 1383- 1390.
  43. Li, Q., et al., Klotho negatively regulated aerobic glycolysis in colorectal cancer via ERK/HIF1α axis. Cell Commun Signal, 2018. 16(1): p. 26.
  44. Wolf, I., et al., Functional variant of KLOTHO: a breast cancer risk modifier among BRCA1 mutation carriers of Ashkenazi origin. Oncogene, 2010. 29(1): p. 26-33.
  45. Liu, C., et al., Klotho gene polymorphisms are related to colorectal cancer susceptibility. Int J Clin Exp Pathol, 2015. 8(6): p. 7446-9.
  46. Xie, B., et al., Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol, 2013. 44(5): p. 795-801.
  47. Pako, J., et al., Assessment of the circulating klotho protein in lung cancer patients. Pathol Oncol Res, 2020. 26(1): p. 233-238.