HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Arsenic sulfide nanoformulation induces erythroid differentiation in chronic myeloid leukemia cells through degradation of BCR-ABL.

AbstractBACKGROUND:
Chronic myeloid leukemia (CML) is a myeloproliferative disorder due to the existence of BCR-ABL fusion protein that allows the cells to keep proliferating uncontrollably. Although tyrosine kinase inhibitors can inhibit the activity of BCR-ABL fusion protein to trigger the cells apoptosis, drug resistance or intolerance exists in part of CML patients. Arsenic sulfide in its raw form (r-As4S4) can be orally administrated and certain therapeutic effects have been found out in the treatment of hematologic malignancies through inducing cell apoptosis.
METHODS:
In this work, a water-dissolvable arsenic sulfide nanoformualtion (ee-As4S4) composed of As4S4 particulates with 470 nm in diameter and encapsulated by a kind of hydrophilic polymer was fabricated and applied to the CML cell line K562, K562/AO2 and primary cells from the bone marrow of CML patients.
RESULTS:
Results showed that instead of inhibiting the activity of BCR-ABL, ee-As4S4 induced direct degradation of BCR-ABL in K562 cells within 6 hr incubation, followed by the occurrence of erythroid differentiation in K562 after 72 hr incubation, evidenced by the significantly upregulated CD235a and benzidine staining, which was not detectable with r-As4S4. The ee-As4S4-induced erythroid differentiation was also observed in K562/AO2 cells and bone marrow mononuclear cells of CML patients. Mechanistic studies indicated that ee-As4S4 induced autophagy by downregulating the level of intracellular ROS and hypoxia-inducible factor-1α significantly, which led to the subsequent degradation of BCR-ABL. When the concentration was increased, ee-As4S4 induced much more significant apoptosis and cell cycle arrest than r-As4S4, and the cytotoxicity of the former was about 178 times of the latter.
CONCLUSION:
ee-As4S4 was capable of inducing significant erythroid differentiation of CML cells by inducing the direct degradation of BCR-ABL; the new effect could improve hematopoietic function of CML patients as well as inhibit the leukemic cell proliferation.
AuthorsTao Wang, Tao Wen, Hongmin Li, Bing Han, Suisui Hao, Chuan Wang, Qiang Ma, Jie Meng, Jian Liu, Haiyan Xu
JournalInternational journal of nanomedicine (Int J Nanomedicine) Vol. 14 Pg. 5581-5594 ( 2019) ISSN: 1178-2013 [Electronic] New Zealand
PMID31413564 (Publication Type: Journal Article)
Chemical References
  • Arsenicals
  • HIF1A protein, human
  • Hypoxia-Inducible Factor 1, alpha Subunit
  • Reactive Oxygen Species
  • Sulfides
  • arsenic trisulfide
  • Fusion Proteins, bcr-abl
  • p38 Mitogen-Activated Protein Kinases
Topics
  • Apoptosis (drug effects)
  • Arsenicals (pharmacology)
  • Autophagy (drug effects)
  • Cell Cycle Checkpoints (drug effects)
  • Cell Differentiation (drug effects)
  • Down-Regulation (drug effects)
  • Drug Compounding
  • Erythroid Cells (cytology, drug effects, ultrastructure)
  • Fusion Proteins, bcr-abl (metabolism)
  • Humans
  • Hypoxia-Inducible Factor 1, alpha Subunit (genetics, metabolism)
  • K562 Cells
  • Leukemia, Myelogenous, Chronic, BCR-ABL Positive (drug therapy, pathology)
  • MAP Kinase Signaling System (drug effects)
  • Nanoparticles (chemistry)
  • Phosphorylation (drug effects)
  • Proteolysis (drug effects)
  • Reactive Oxygen Species (metabolism)
  • Sulfides (pharmacology)
  • p38 Mitogen-Activated Protein Kinases (metabolism)

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: