scholarly journals Human Wharton’s Jelly Stem Cell Secretions Inhibit Human Leukemic Cell Line K562 in vitro by Inducing Cell Cycle Arrest and Apoptosis

Author(s):  
Muneerah A. H. Huwaikem ◽  
Gauthaman Kalamegam ◽  
Ghadeer Alrefaei ◽  
Farid Ahmed ◽  
Roaa Kadam ◽  
...  

Emerging resistance to the tyrosine kinase inhibitors that target the BCR-ABL1 oncoprotein has prompted research for novel therapeutics against chronic myeloid leukemia (CML). Herein, we evaluated the tumor inhibitory properties of the human Wharton’s jelly stem cells (hWJSCs) co-culture (hWJSC-CC) and their extracts, namely, the hWJSC-conditioned medium (hWJSC-CM; 100%) and hWJSC-lysate (hWJSC-L; 15 μg/ml), on a CML cell line K562 in vitro. The hWJSCs expressed mesenchymal stem cell (MSC)-related cluster of differentiation (CD) markers and demonstrated mesodermal tissue differentiation potential. The cell metabolic activity showed a mean maximal decrease in the K562 cells by 49.12, 41.98, and 68.80% following treatment with the hWJSC-CC, hWJSC-CM, and hWJSC-L, respectively, at 72 h. The sub-G1 population in the cell cycle was decreased by 3.2, 4.5, and 3.8% following treatment with the hWJSC-CC, hWJSC-CM, and hWJSC-L, whereas the G2/M cell population was increased by 13.7 and 12.5% with the hWJSC-CM and hWJSC-L, respectively, at 48 h. Annexin V–allophycocyanin (APC) assay showed an increase in the apoptotic cells by 4.0, 3.9, and 4.5% at 48 h. The expression of pro-apoptotic BAX and CASP3 genes were increased, whereas BIRC5 (Survivin) was decreased compared with the control. The pro-inflammation-related genes, namely, IFN-γ, TNF-α, IL-1β, IL-6, IL-8, and IL-12A, were decreased, whereas the anti-inflammatory genes, namely, IL-4 and IL-10, were increased following treatment with the hWJSC-CC, hWJSC-CM, and hWJSC-L at 48 h. Multiplex bead-based cytokine assay also demonstrated decreases in the pro-inflammatory cytokines (IFN-γ, TNF-α, IL-1β, IL-6, and IL-12) and an increase in the anti-inflammatory cytokine (IL-10) compared with the control. The pro-inflammatory cytokine IL-8 showed an increase with the hWJSC-CC and decreases with both the hWJSC-CM and the hWJSC-L. The hWJSCs and their extracts inhibited the K562 cells by causing cell cycle arrest and inducing apoptosis via the soluble cellular factors. However, an in vivo evaluation is necessary to unravel the true potential of the hWJSCs and their extracts before its use in CML inhibition.

Biomaterials ◽  
2012 ◽  
Vol 33 (26) ◽  
pp. 6162-6171 ◽  
Author(s):  
Yan-Qing Guan ◽  
Zhibin Li ◽  
Aini Yang ◽  
Zheng Huang ◽  
Zhe Zheng ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5004-5004
Author(s):  
Yuliya Linhares ◽  
Jade Dardine ◽  
Siavash Kurdistani

Abstract Abstract 5004 Introduction: Amiloride is an FDA approved potassium-sparing diuretic which targets Na+/H+ exchanger isoform 1 (NHE1). NHE1 is responsible for the regulation of the intracellular pH, as well as cell-cycle and apoptosis. In supra-pharmacologic concentrations, amiloride non-specifically inhibits protein kinases. Recent study demonstrated that proapoptotic effect of amiloride in CML cell lines is linked to the modulation of the alternative splicing of Bcl-x, HIPK3, and BCR/ABL genes and is independent of pHi. Here, we demonstrate that pharmacologic doses of amiloride preferentially induce growth inhibition, cell cycle arrest and apoptosis in Flt3-ITD positive acute myeloid leukemia cell lines as compared to Bcr-Abl positive leukemia cell line. Our data suggests that amiloride may have an effect on Flt3 signaling and that its treatment potential for Flt3-ITD positive acute myeloid leukemia needs to be explored. Methods: MV4-11, MOLM13 and K562 cells lines in log-phase growth were used for the experiments. Analysis of the baseline Flt3 expression and phosphorylation status was assessed via Flt3 immunoprecipitation and Western blotting for Flt3 and phosphotyrosine. Cells were incubated with various amiloride concentrations; equal volume dilutions of DMSO were used for control. Cell counting and trypan blue exclusion viability was performed on TC10 Bio-Rad automated cell counter. The cell cycle analysis was performed applying propidium iodide staining. To assess for apoptosis and cell death, we used annexin V/PI staining kit and flow cytometry. Results: MOLM13 and MV4-11 cell lines carry activating Flt3-ITD mutation. We confirmed the expression and constituative activation of Flt3 in MOLM13 and MV4-11 cells with Western blotting. Flt3 protein was not detectable in K562 cell line. Amiloride at 0.025 mM and 0.05 mM completely inhibited the growth of MV4-11 cells after 24 hrs of treatment with no significant increase in total or live cell numbers at 72 hrs, but only mildly affected K562 cell proliferation. While the above amiloride concentrations caused cell death in MV4-11 and MOLM13 cell lines, there was no increased cell death in K562 cells. Incubation of MOLM13 and MV4-11 cell lines with 0.05 mM amiloride for 20 hrs induced cell cycle arrest. In MV4-11 cell line, the proportion of S phase cells after amiloride treatment was 15.4% (SD=5.4%) as compared to 31.3% (SD=1.4%) in control. MOLM13 cell line demonstrated 15.3% (SD=4.7%) of cells in S after amiloride treatment as compared to 35.3% (SD=2.4%) cells in S phase in control treatment. In K562 cell line, there was less effect with 52% (SD=4.2%) of cells in S phase in control as compared to 37% (SD=8.9%) in amiloride treatment. MV4-11 and MOLM 13 cell lines were more sensitive than K562 cells to amiloride induced apoptosis with 28.8% (control 12.7%) of MV4-11 cells, 11.4% (control 7.4%) of MOLM13 cells, and 11.4% (control 8.6%) of K562 cells being apoptotic after 20 hr treatment with 0.05mM amiloride. At 72 hrs of amiloride treatment 34% (control 1.5%) of MV4-11 cells, 17% (control 5%) of MOLM13 cells and 11% of K562 cells (control 8.9%) were apoptotic. Amiloride had similar effect on the number of dead cells with no increase in total cell death in K562 cell line. Upon treatment with increasing amiloride concentrations, there was dose-dependent increase in cell death and apoptosis in all three cell lines with K562 line showing relative resistance to amiloride. Discussion: Our results demonstrate that amiloride induces cell cycle arrest and inhibits proliferation of Flt3-ITD positive cell lines MV4-11 and MOLM13 as well as K562 cell line at a pharmacologic concentration of 0.05 mM. Both, cell cycle arrest and antiproliferative effect are more pronounced in Flt3-ITD positive cells lines while it is mild in Bcr-Abl positive K562 cell line. Pharmacologic doses of amiloride induce cell death and apoptosis in Flt3-ITD positive cell lines but not in K562 cell line. Both, Bcr-Abl and Flt3 signaling stimulates proliferation and inhibits apoptosis in myeloid leukemia cells. Our study suggests that amiloride may induce cell cycle arrest and apoptosis via modulating Flt3 signaling cascade. We are currently investigating the effects of amiloride on Flt3 phosphorylation. In conclusion, our data suggests that amiloride presents a potential treatment option for Flt3-ITD positive acute myeloid leukemia. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 2019 ◽  
pp. 1-7
Author(s):  
Xiaolong Du ◽  
Yujing Zhao ◽  
Yongqian Ma ◽  
Hongshun Xing ◽  
Xingang Li

Objective. To investigate the possible development of radix hedysari polysaccharide as an antiglioma drug, we studied the effect of radix hedysari polysaccharide on glioma cells in vitro and the growth of glioma in nude mice and on the phagocytosis of macrophages in nude mice with glioma. Methods. The effect of radix hedysari polysaccharide on the growth of glioma was studied based on U251 cell line in vitro. The effect of radix hedysari polysaccharide on the growth of glioma was studied in vivo. The growth inhibition rate of radix hedysari polysaccharide on U251 cell line was determined by the MTT assay. The cell cycle of U251 was analyzed by flow cytometry. The expression of cytokines in U251 cells and tumor tissues was detected using PCR. The phagocytosis of macrophages in the serum of glioma nude mice was detected by Giemsa staining. TNF-α signaling pathway proteins in the serum of glioma nude mice were detected by ELISA. Results. Radix hedysari polysaccharide inhibited the growth of U251 cells, induced apoptosis in G1 phase by cell cycle arrest, and facilitated apoptosis in glioma mice by regulating cell cycle. Mice injected with radix hedysari polysaccharide showed delayed tumor growth and grew slowly. Radix hedysari polysaccharide enhanced the phagocytosis of macrophages in glioma nude mice. Radix hedysari polysaccharides could inhibit tumor development by regulating the immune function of tumor mice and affecting the TNF-α signaling pathway. Conclusion. Radix hedysari polysaccharide can effectively inhibit the growth of glioma and affect the TNF-α signaling pathway, thus playing an antiglioma role.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4301-4301
Author(s):  
Yuko Mishima ◽  
Yasuhito Terui ◽  
Yuji Mishima ◽  
Toshihiro Takizawa ◽  
Shinya Kimura ◽  
...  

Abstract Back ground; Accumulating evidence suggests that programmed cell death is not defined as apoptosis but cells use different pathways for active self-destruction as reflected by different morphology. Autophagic cell death (APCD), which also be designated type II programmed cell death and appears to be a phylogenetically old phenomenon, is observed in physiological and disease state. Indeed, APCD is observed in several neurodegenerative diseases and cancer cells, however, the APCD in leukemic cells has not been reported yet. Here, we found APCD was occurred in bcr-abl-expressing CML cell lines. Methods; Bcr-abl-positive cell line, K562, were cultured with 2nM of TPA up to 48hours. Subsequently, we examined the morphological change by light microscopy and electronmicroscopy. To evaluate the APCD, TPA-treated K562 cells were labeled with autofluorescent agent Monodansylcadaverin(MDC), which specifically accumulates in autophagosomes and we measured accumulation of autophagosmes in cytoplasm by fluorescent microplate reader. TPA continuous activated MAPK of K562 cells and induced cell cycle arrest in G0/G1 phase. In order to elucidate the role of activation of MAPK in APCD, MAPK in K562 cells were inactivated by MAPK inhibitor, U0126 or dominant negative MAPK-transfection, and then, the cells were treated with TPA. Furthermore, to evaluate the involvement of cell cycle arrest in APCD, bcr-abl-transfected murine leukemic cell line, BAF3, was treated with cell cycle arrest inducer, mimosine. In addition, we investigated the expression of an autophagy-related molecule, Beclin 1 by immunohistochemical analysis. Result; The 48 hours’ treatment with TPA induced cell death in K562 cells, which accumulated plenty of autophagosomes in cytoplasm. MDC labeling assay revealed that accumulation of autophagosomes were increased in time- and dose-dependent manner. On the other hand, MAPK-inactivated K562 cells exhibited resistance to TPA-induced APCD. Furthermore, the bcr-abl-transfected BAF3 cells exhibited continuous activation of MAPK and underwent APCD by cell cycle arrest by mimosine. Immunohistochemical analysis revealed that Beclin 1 shuttled between nuclear and cytoplasms and co-localized with MAPK during APCD progression. Conclusion: APCD in bcr-abl-positive leukemia cells were closely involved in G0/G1 cell cycle arrest and continuous activation of MAPK. These findings are useful in developing the novel strategy to treatment of Ph1 positive leukemia via alternative pathways and another type of cell death.


2019 ◽  
Vol 18 (11) ◽  
pp. 1563-1572 ◽  
Author(s):  
Nimmy Kumar ◽  
Akhila H. Shrungeswara ◽  
Sanchari B. Mallik ◽  
Subhankar Biswas ◽  
Jesil Mathew ◽  
...  

Background: Hepatocellular carcinoma (HCC) is the fifth leading cause of cáncer mortality. Elytranthe parasitica (L.) Danser (EP), a hemiparasitic plant (Loranthaceae) has potent anti-cancer properties. Objective: In the study, we investigated the effect of EP fractions on the expression of apoptosis and mitogenactivated protein kinase (MAPK) markers deregulated in HCC. Bioactivity fractionation was performed to isolate the phytochemical(s) exerting anti-tumor activity in HepG2 cells. Method: Anti-proliferative, clonogenic and anti-metastatic effects of EP fractions were examined in hepatocellular carcinoma cell line, HepG2 by Sulphorhodamine B, colony formation and scratch wound assays respectively in hepatocellular cell line, HepG2. The effects of EP fractions on key markers of apoptosis and MAPK signaling pathways were explored. </P><P> Key findings: EP bioactive fractions showed significant anti-tumor potential, reduced clonogenicity and considerably inhibited cell migration in HepG2 cells in vitro. The fractions augmented annexin V binding and induced apoptosis by causing cell cycle arrest at G2/M and S phase checkpoints. The fractions increased expression levels of p53, bad, cleaved PARP (Poly ADP ribose polymerase) and cleaved Caspase-3. Expression levels of phosphorylated ERK1/2 (Extracellular signal-regulated kinase) were downregulated. Pinocembrin-7-O-ß-D-glucoside and chrysin were isolated and characterized for the first time from Elytranthe parasitica (L.) Danser. Conclusion: Our findings reveal that EP fractions induced cell cycle arrest and triggered apoptosis in HepG2 cells by upregulating apoptosis and deactivating MAPK pathway. It signifies that pinocembrin glycoside and chrysin are bioactive phytochemicals contributing to the potent anti-hepatocarcinoma effects on HepG2 cells. Hence, bioactive EP fractions could be used as a therapeutic agent for effective HCC therapy.


2013 ◽  
Vol 2013 ◽  
pp. 1-6 ◽  
Author(s):  
Mostafa A. Borahay ◽  
Fangxian Lu ◽  
Bulent Ozpolat ◽  
Ibrahim Tekedereli ◽  
Bilgin Gurates ◽  
...  

Objective. To determine the effects of Mullerian inhibiting substance (MIS) treatment on endometriosis cells through study of apoptosis and autophagy. Design. Experimental in vitro study. Setting. University research laboratory. Cell Line. CRL-7566 endometriosis cell line. This line was established from a benign ovarian cyst taken from a patient with endometriosis. Interventions. In vitro treatment with MIS. Main Outcome Measures. The main outcome measures were cellular viability, proliferation, cell-cycle arrest, and induction of apoptosis and autophagy in endometriotic cells. Results. MIS treatment inhibited proliferation of endometriosis cells and induced apoptosis, as indicated by Annexin V staining, and induced caspase-9 cleavage and cell-cycle arrest, as evidenced by increased expression of p27 CDK-inhibitor. MIS treatment also induced autophagy in endometriosis cells as demonstrated by a significant increase in LC3-II induction, a hallmark of autophagy. Conclusions. MIS inhibits cell growth and induces autophagy, as well as apoptosis, in ectopic endometrial cell lines. Our results suggest that MIS may have a potential as a novel approach for medical treatment of endometriosis. Further studies may be needed to test the efficacy of MIS treatment in animal models and to develop MIS treatment specifically targeted to the endometriosis.


2018 ◽  
Vol 6 (2) ◽  
pp. 327-336 ◽  
Author(s):  
Wuya Chen ◽  
Wenwen Wang ◽  
Liyi Chen ◽  
Jiamei Chen ◽  
Xinhua Lu ◽  
...  

For inducing long-term G1-phase cell cycle arrest, co-immobilized TNF-α plus IFN-γ polymeric drugs are very efficient tools that inhibit cervical cancer.


2018 ◽  
Vol 46 (1) ◽  
pp. 381-390 ◽  
Author(s):  
Zahraa R. Shamsee ◽  
Ali Z. Al-Saffar ◽  
Ahmed F. Al-Shanon ◽  
Jameel R. Al-Obaidi

Sign in / Sign up

Export Citation Format

Share Document