scholarly journals Punicalagin promotes autophagy to protect primary human syncytiotrophoblasts from apoptosis

Reproduction ◽  
2016 ◽  
Vol 151 (2) ◽  
pp. 97-104 ◽  
Author(s):  
Ying Wang ◽  
Baosheng Chen ◽  
Mark S Longtine ◽  
D Michael Nelson

Punicalagin is a prominent polyphenol in pomegranate juice that protects cultured syncytiotrophoblasts from stress-induced apoptosis. Here, we test the hypothesis that punicalagin has this effect by inhibiting the mTOR kinase pathway to enhance autophagic turnover and limit apoptosis in cultured primary human syncytiotrophoblasts. In syncytiotrophoblasts, starvation, rapamycin, or punicalagin all decreased the expression of phosphorylated ribosomal protein S6, a downstream target of the mTOR kinase, and of the autophagy markers, LC3-II and p62. In contrast, in the presence of bafilomycin, an inhibitor of late stages of autophagy and degradation in the autophagolysosome, syncytiotrophoblasts exposed to starvation, rapamycin, or punicalagin all showed increased levels of LC3-II and p62. The number of LC3-II punctae also increased in punicalagin-treated syncytiotrophoblasts exposed to chloroquine, another inhibitor of autophagic degradation, and punicalagin increased the number of lysosomes. The apoptosis-reducing effect of punicalagin was attenuated by inhibition of autophagy using bafilomycin or knockdown of the autophagy related gene,ATG16L1. Collectively, these data support the hypothesis that punicalagin modulates the crosstalk between autophagy and apoptosis to promote survival in cultured syncytiotrophoblasts.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2474-2474
Author(s):  
Piotr Smolewski ◽  
Agnieszka Janus ◽  
Barbara Cebula ◽  
Anna Linke ◽  
Krzysztof Jamroziak ◽  
...  

Abstract Background: Rapamycin (RAPA) is an inhibitor of mTOR kinase pathway. In vitro low doses of this agent induce cell cycle arrest in G1 phase, whereas higher concentrations of RAPA exert proapoptotic effects. Aim: We assessed cytotoxicity of RAPA alone or in combination with cytarabine (cytosine arabinoside, ARA-C) in acute myeloblastic leukemia (AML) cells and in normal lymphocytes obtained from 10 healthy volunteers. Methods: AML cells (in vitro HL-60 cell line and ex vivo leukemic cells) and phytohemaglutynin (PHA)-stimulated normal lymphocytes were treated for 24 – 48 h with 1 ng/ml RAPA alone or in combination with 50 nM cytarabine (Ara-C). Moreover, cells was pre-incubated with RAPA for 24 h and then Ara-C was added for the next 24 h. Untreated cultures and those treated with RAPA, Ara-C or PHA alone served as respective controls. The proapoptotic effect was assessed by Annexin V assay and presented as a percentage of Annexin-V-positive cells (apoptotic index; AI). Cell cycle was analyzed by DNA distribution in propydium iodide/RN-ase stained cells. Cyclin D3, A and E expression was also measured using flow cytometry. Results: Median AI induced in HL-60 cells after 24 h treatment with RAPA+Ara-C (30.1%) was significantly higher than induced by RAPA (7.2%) or Ara-C (18.5%) alone (p=0.002 and p=0.03, respectively). The RAPA+Ara-C combination exerted additive effect (combination index 0.87) in that model. Additional 24 hour pretreatment with RAPA further increased apoptosis (median AI 41.5%, vs. 10.9% after 48 h-RAPA alone). In contrast to leukemic cells, pretreatment of normal PHA-stimulated lymphocytes with RAPA caused their G1 phase cell cycle arrest, with significant decrease in cyclin D3 expression (vs. untreated cells - p<0.001). This resulted in prevention of Ara-C-induced cytotoxicity in healthy lymphocytes, when Ara-C was added for another 24 h. Importantly, that protective effect was reversible when RAPA-treated lymphocytes were rinsed and then cultured in fresh, RAPA-free medium for the next 24 h. In another set of experiments, cells from 12 de novo AML patients were treated with RAPA and Ara-C in above concentrations and time settings. RAPA and Ara-C were administrated to isolated peripheral blood mononuclear cells (PBMC). PBMC were immunophenotyped before and after treatment. Leukemic blasts were marked for individually chosen antigen, most characteristic for leukemic clone in particular patient. Normal CD3+ lymphocytes were also detected. Finally, Annexin V staining was performed. Based on that simultaneous three-color staining the proapoptotic effects of treatment could be measured by flow cytometry in both leukemic blasts and normal CD3+ cells. Thus, we found that pretreatment with RAPA protected majority of CD3+ cells (median of alive cells 85.5%) from Ara-C-induced apoptosis, whereas the leukemic blasts AI was higher than in samples treated with Ara-C. After Ara-C alone CD3+ rate decreased significantly (median 35.1%). Conclusions: Pretreatment with RAPA enhances cytotoxic effect of Ara-C on leukemic cells, but not on healthy lymphocytes. The phenomenon is probably due to reversible arrest of healthy cells in G1 phase of cell cycle by low doses of RAPA, what causes their transient resistance to proapoptotic action of cytostatic drugs. In contrast, the same RAPA doses selectively sensitizes leukemic cells to cytostatics. This suggests, that inhibition of mTOR kinase prior to cytostatics administration may result in selective anti-tumor treatment, with protection of normal cells.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3104-3104 ◽  
Author(s):  
Nicholas Burwick ◽  
Jeffrey J Delrow ◽  
Akiko Shimamura

Abstract Dexamethasone has been used to treat Multiple Myeloma (MM), a plasma cell cancer, for over forty years. Nonetheless, dexamethasone toxicity is a source of morbidity for many patients and drug resistance invariably develops. A better understanding of the mechanisms of dexamethasone activity could provide novel avenues for drug development. To gain new insights into dex activity in MM, we started with the clinical observation that dex is an effective treatment for both MM and the ribosomal disorder Diamond Blackfan anemia (DBA). Interestingly, dex has opposite effects in these two disorders, promoting apoptosis in MM and inducing an erythroid response in DBA. Given these opposing responses to dex, we hypothesized that dex disrupts ribosomal profiles in MM, altering the translation of specific mRNAs to promote apoptosis. To address this hypothesis, we performed an unbiased assessment of mRNA translation in MM +/- dex, using translational (polysome) profiling. We show that dex disrupts ribosomal profiles in sensitive (MM1.S), but not resistant (MM1.R) myeloma cell lines as early as 4 hours, well-preceding dex-induced apoptosis. These effects are dose and time dependent. We next treated the MM1.S and MM1.R cell lines with 1μM dex or control for 4 hours. Cells were lysed and separated by ultracentrifugation. Polysome gradients were obtained, and the gradients were fractionated into two pools: (A) non-polysome associated RNA and (B) polysome associated RNA. Pool B is enriched for actively translated mRNA. A translation coefficient for each condition was defined as the amount of mRNA in polysome/non-polysome pools. Unfractionated total cellular RNA was used to assess transcriptional changes. RNA was analyzed using human gene arrays. The results demonstrate that dex alters a group of translational networks that are distinct from its effect on transcriptional networks. The top translational network modulated by dex was eIF2 signaling (p-value 1.4 E-09). The eIF2 kinase pathway regulates protein translation, and repression of this pathway paradoxically increases the translation of the transcription factor ATF4. We show that dex increases ATF4 translation. In addition dex up-regulates transcription of the ATF4 target gene DDIT4 (REDD1). Dex also represses the translation of ribosomal protein genes downstream of REDD1, including RPS6, RPS19 and RPS24. The large ribosomal protein genes RPL3 and RPL10 were unchanged, suggesting that dex represses the translation of a subset of ribosomal protein genes. Changes in protein expression were validated by immunoblot. Finally, we tested whether specific inhibition of the eIF2 kinase pathway could overcome dex resistance. Using an MTS assay we demonstrate that BTdCPU, a specific eIF2 kinase inhibitor, induces cytotoxicity in dex sensitive (MM1.S, RPMI8266) and dex resistant (MM1.R, U266) cell lines. Using flow cytometry, BTdCPU also induces apoptosis in both dex sensitive and resistant cell lines. Furthermore, BTdCPU and dex have additive cytotoxicity/apoptosis in the MM1.S cell line. Lastly, we show that BTdCPU specifically up-regulates the ATF4 target genes REDD1 and CHOP. Up-regulation of the pro-apoptotic gene CHOP by BTdCPU may explain the additive cytotoxicity in conjunction with dex. In summary, 1) The eIF2 kinase pathway is modulated by dexamethasone at the level of mRNA translation in multiple myeloma. 2) Direct targeting of the eIF2 kinase pathway with BTdCPU overcomes dex resistance. We show that dexamethasone alters the translation of specific pathways in MM (independent of transcription), including eIF2, revealing potential new avenues for drug development. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Kehan Li ◽  
Cunte Chen ◽  
Rili Gao ◽  
Xibao Yu ◽  
Youxue Huang ◽  
...  

AbstractT-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of leukemia with poor prognosis, and biomarkers and novel therapeutic targets are urgently needed for this disease. Our previous studies have found that inhibition of the B-cell leukemia/lymphoma 11B (BCL11B) gene could significantly promote the apoptosis and growth retardation of T-ALL cells, but the molecular mechanism underlying this effect remains unclear. This study intends to investigate genes downstream of BCL11B and further explore its function in T-ALL cells. We found that PTK7 was a potential downstream target of BCL11B in T-ALL. Compared with the healthy individuals (HIs), PTK7 was overexpressed in T-ALL cells, and BCL11B expression was positively correlated with PTK7 expression. Importantly, BCL11B knockdown reduced PTK7 expression in T-ALL cells. Similar to the effects of BCL11B silencing, downregulation of PTK7 inhibited cell proliferation and induced apoptosis in Molt-4 cells via up-regulating the expression of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p27. Altogether, our studies suggest that PTK7 is a potential downstream target of BCL11B, and downregulation of PTK7 expression via inhibition of the BCL11B pathway induces growth retardation and apoptosis in T-ALL cells.


1977 ◽  
Vol 252 (17) ◽  
pp. 6217-6221
Author(s):  
M A Treloar ◽  
M E Treloar ◽  
R Kisilevsky

Sign in / Sign up

Export Citation Format

Share Document