scholarly journals Exploring NCX4040, an Aspirin Derivative, as a Potential Treatment for Benign Prostatic Hyperplasia

2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A765-A765
Author(s):  
Preksha Vaibhav Shahagadkar ◽  
Gnanasekar Munirathinam

Abstract Benign Prostatic hyperplasia (BPH) is a leading cause of lower urinary tract symptoms which affects men above 50 years of age. Chronic inflammation and abnormal proliferation of stromal and epithelial cells are implicated in BPH disease onset. The symptoms of BPH include back pain and difficulty in emptying bladder. Finasteride, sildenafil and tamsulosin are some of the drugs used to ease difficulty urinating and relax the muscles of the gland. Transurethral resection of the prostate or laser surgery can be performed to treat severe symptoms. However, these therapies have deleterious effects such as low blood pressure, ejaculatory dysfunction, and lump formation. Hence, there is an unmet need for potential drugs against BPH. Nonsteroidal anti-inflammatory drugs (NSAIDs) have proven to be effective in cancers but their applicability in BPH condition is yet to be fully explored. Aspirin, one of the NDSAIDs, has anti-tumor and anti-inflammatory properties at higher doses. NCX4040, a nitric oxide releasing derivative of aspirin, could prove to be effective against BPH, since it can inhibit abnormal cell proliferation and serve as a vasodilator. We hypothesize that NCX4040 would be an effective drug to treat BPH. BPH-1 epithelial and WPMY-1 stromal cells were used as in vitro models of BPH. MTT assay was performed to check the inhibitory effect of NCX4040 and blocking agents like catalase and N-acetyl-L-cysteine (NAC) were explored on cells after treatment. Clonogenic assay was done to explore the colony formation ability of cells. Spheroid assay was performed to analyze the anti-proliferative effect of NCX4040. Annexin V/PI and cell cycle analysis was performed to check for apoptosis and cell cycle arrest in the cells. Western blot was done to assess the signaling molecules altered by NCX4040 in BPH-1 cells. Confocal immunofluorescence was employed to analyze the dynamics of actin filaments after treatment in cells. Our studies revealed that NCX4040 inhibited the cell viability of BPH-1 and WPMY-1 in a dose dependent manner with IC50 predicted at 5µM and 2.5µM respectively. Of note, catalase and NAC blocked the effect of NCX4040 on prostate cells. Colony formation assay result implied a gradual decrease in the number of colonies of cells treated with NCX4040 with 2.5µM and 5µM doses. Spheroid assay in BPH-1 cells showed inhibitory effects after treatment. Cell cycle analysis by flowcytometry inferred that cell cycle arrest at G2/M phase and annexin V analysis indicated that activation of apoptosis in cells following treatment. Phalloidin staining showed decrease in the actin filament intensity in cells. At the molecular level, NCX4040 downregulated the expression of key markers such as RhoA, p65, COX-2, PCNA, Cyclin D3, and PDE-5 in BPH-1 cells. Taken together, NCX4040 could be used as a potential agent to manage BPH with minimal side effects, which needs further evaluation in animal models.

In Vivo ◽  
2018 ◽  
Vol 32 (6) ◽  
pp. 1373-1379 ◽  
Author(s):  
LIANG-CHIH LIU ◽  
YUNG-HSIANG LIN ◽  
YING-CHAO LIN ◽  
CHI-TANG HO ◽  
CHAO-MING HUNG ◽  
...  

2021 ◽  
Author(s):  
Xiaoyue Chen ◽  
Jingrui Song ◽  
Dongbo Yuan ◽  
Qing Rao ◽  
Kehua Jiang ◽  
...  

Benign prostatic hyperplasia (BPH) is a common disease that occurs mainly in older men. The pathogenesis of BPH is complex and patients face a prolonged treatment course, and novel drugs with better selectivity and lower toxicity are required. incaspitolide A (compound TMJ-12) is a germacrane-type sesquiterpenoid compound extracted from the plant Carpesium carnuum. Extracts of C. carnuum are known to exert suppressive effects on BPH-1 cells. In this study, we investigated the molecular mechanisms underlying the suppressive effect of TMJ-12 specifically on BPH-1 cells. A cytotoxicity assay indicated that TMJ-12 inhibited BPH-1 cell proliferation, while flow cytometry assays showed that TMJ-12 induced G2/M phase cell cycle arrest and the apoptosis of BPH-1 cells. TMJ-12 was also shown to regulate the expression of several apoptosis- and cell cycle-related proteins, namely Bcl-2, Bax, Bad, Caspase-9, Caspase-3, CDK1, Cyclin B1, CDC25C, and c-Myc, among others. Collapse of the mitochondrial membrane potential (ΔΨm) following exposure to TMJ-12 was detected with the JC-1 staining assay. Further investigation revealed that treatment with TMJ-12 inhibited the PI3K/AKT pathway by increasing the expression of PTEN. Taken together, the results suggest that TMJ-12 prevents BPH-1 cell proliferation via the PI3K/AKT pathway by inducing apoptosis and cell cycle arrest.


Author(s):  
Agata Kabała-Dzik ◽  
Anna Rzepecka-Stojko ◽  
Robert Kubina ◽  
Żaneta Jastrzębska-Stojko ◽  
Rafał Stojko ◽  
...  

1) Background: Studies indicate that caffeic acid (CA), caffeic acid phenethyl ester (CAPE) are compounds with potent chemopreventive effects. Breast cancer is a common cancer among women worldwide. The study shows comparison of caffeic acid and its ester activity in the cells of breast cancer line MDA-MB-231; 2) Methods: The cells of MDA-MB-231 were treated by CA and CAPE with doses from 10 to 100 µM in time 24 h and 48 h. Cytotoxicity MTT test, apoptosis by Annexin V and cell cycle with Dead Cell Assay were performed; 3) Results: The cytotoxic activity was greater for CAPE comparing to CA, in both incubation time (same dosage). IC50 values for CAPE were 27.84 (24h) and 15.83 (48h) and >10000 (24h) and >1000 (48h) for CA. Polyphenols induced apoptosis, higher apoptotic effect observed for CAPE (dose dependent). CAPE induced cell cycle arrest in S phase (time and dose dependent). Dose dependent decline G0/G1 phase (48h) and elimination of phase G2/M (100 µM of CAPE). For CA, only after 48 hours, small effect of cell cycle at phase S (however dose dependent), and slight decline of phase G0/G1 and G2/M only for highest doses (50 and 100 µM); 4) Conclusions: Comparing CA and CAPE activity, on the MDA-MB-231, we clearly see better activity of CAPE, with the same dosage and experiment time.


2020 ◽  
Author(s):  
Yu Liu ◽  
Risheng Huang ◽  
Deyao Xie ◽  
Xiaoming Lin ◽  
Liangcheng Zheng

Abstract Background: ZNF674-AS1, a recently characterized long noncoding RNA, shows prognostic significance in hepatocellualar carcinoma and glioma. However, the expression and function of ZNF674-AS1 in non-small cell lung cancer (NSCLC) is unclear. Methods: In this work, we investigated the expression of ZNF674-AS1 in 83 pairs of NSCLC specimens and adjacent noncancerous lung tissues. The clinical significance of ZNF674-AS1 in NSCLC was analyzed. The role of ZNF674-AS1 in NSCLC growth and cell cycle progression was explored. Results: Our data show that ZNF674-AS1 expression is decreased in NSCLC compared to normal tissues. ZNF674-AS1 downregulation is significantly correlated with advanced TNM stage and decreased overall survival of NSCLC patients. Overexpression of ZNF674-AS1 inhibits NSCLC cell proliferation, colony formation, and tumorigenesis, which is accompanied by a G0/G1 cell cycle arrest. Conversely, knockdown of ZNF674-AS1 enhances the proliferation and colony formation of NSCLC cells. Biochemically, ZNF674-AS1 overexpression increases the expression of p21 through downregulation of miR-423-3p. Knockdown of p21 or overexpression of miR-423-3p blocks ZNF674-AS1-mediated growth suppression and G0/G1 cell cycle arrest. In addition, ZNF674-AS1 expression is negatively correlated with miR-423-3p in NSCLC specimens. Conclusions: ZNF674-AS1 suppresses NSCLC growth by downregulating miR-423-3p and inducing p21. This work suggests the therapeutic potential of ZNF674-AS1 in the treatment of NSCLC.


2019 ◽  
Author(s):  
I Wayan - Suardana ◽  
I Gusti Ngurah - Sudisma ◽  
Komang Januartha Putra Pinatih ◽  
Dyah Ayu Widiasih

Abstract Background Apoptosis and cell cycle arrest induction are targeted in the strategy of cancer therapy. Furthermore, bacterial toxins such as Shiga-like toxin producing Escherichia coli have been suggested to be used as a novel therapeutic agent against tumor malignancies, either as independent anti-neoplastic agents, or in combination treatment with chemo or radiotherapy. The aim of study was to investigate the potency of Shiga-like toxin originating from local strains of E. coli O157:H7 which was known less toxic than ATCC 43894 as a new cancer therapy. Methods As many as 10 culture cells T47D cell line were subjected by crude extract Shiga-like toxin originating from five local isolates of E. coli O157:H7 with each codes KL-48(2), SM-25(1), SM-7(1), DS-21(4), and one isolate ATCC 43894 as a control with IC50 doses, respectively. The treatment was observed for 24 h, with two replications. An FITC-Annexin V and PI assay was used to observe apoptosis and necrosis effect, and simultaneously with cell cycle analysis using propidium iodide (PI) staining. Results The study shown that T47D cells treated with Shiga-like toxin from local strain KL-48 (2) show the lowest viable cell, followed by SM7(1), ATCC 43894, SM-25(1), DS-21(4) in contrary with the control cells with each percentages at 15.20, 16.36, 22.17, 22.64, 33.86, and 94.36%, respectively. The results were also confirmed by the induction of the cell cycle arrest in phase G0-G1 as inactive phase, i.e. 66.41, 63.37, 61.52, 55.36 and 47.28% for T47D cells treated with toxins of KL-48(2), ATCC 43894, SM 25(1), SM 7(1), and DS 21(4), respectively. Conclusions These results show tendency deleterious effect of Shiga-like toxin from local isolates on T47D cells, so It is concluded that they have potency as a good anticancer drug against Gb3-expressing breast cancer.


2020 ◽  
Vol 19 ◽  
pp. 153473541990116 ◽  
Author(s):  
Ali H. El-Far ◽  
Noureldien H. E. Darwish ◽  
Shaker A. Mousa

Cellular senescence is a process of physiological growth arrest that can be induced by intrinsic or extrinsic stress signals. Some cancer therapies are associated with senescence of cancer cells with a typical cell cycle arrest. Doxorubicin (Dox) induces senescence by a p53-dependent pathway and telomere dysfunction of numerous cancers. However, cellular senescence induces suppression in proliferation activity, and these cells will remain metabolically active and play an important role in tumor relapse and development of drug resistance. In the current study, we investigated the apoptotic effect of curcumin (Cur), caffeine (Caff), and thymoquinone (TQ) on senescent colon cancer HCT116 and breast cancer MCF7 cell lines treated with Dox. Results showed typical senescence markers including decreased bromodeoxyuridine incorporation, increased accumulation of senescence-associated β-galactosidase (SA-β-gal), cell cycle arrest, and upregulation of p53, P-p53, and p21 proteins. Annexin-V analysis by flow cytometry revealed 2- to 6-fold increases in annexin-V–positive cells in Dox-treated MCF7 and HCT116 cells by Cur (15 µM), Caff (10 mM), and TQ (50 µM; P < .001). In comparison between proliferative and senescent of either HCT116 or MCF7 cells, Caff at 15 mM and TQ at 25 µM induced significant increases in apoptosis of Dox-treated cells compared with proliferative cells ( P < .001). Data revealed that Cur, Caff, and TQ potentially induced apoptosis of both proliferative and senescent HCT116 and MCF7 cells. In vivo and clinical trials are of great importance to validate this result.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1706-1706
Author(s):  
Kenneth H Shain ◽  
Danielle Yarde ◽  
Mark Mead ◽  
Lori Hazlehurst ◽  
William S Dalton

Abstract Multiple Myeloma (MM) is a B cell malignancy characterized by the monoclonal expansion of plasma cells. Although numerous genetic alterations have been implicated in MM pathogenesis, it is widely hypothesized that the bone marrow (BM) microenvironment contributes to MM cell pathogenesis. The BM microenvironmental components, interleukin (IL)-6 and fibronectin (FN), have individually been shown to influence the proliferation and survival of MM cells; however, in vivo these effectors most likely work together. We examined signaling events, cell cycle progression, and levels of drug response in MM cells either adhered to FN via β1 integrins, stimulated with IL-6, or with the two combined. IL-6 and FN adhesion have been demonstrated to protect cells from a host of cytotoxic stimuli suggesting co-stimulation of MM cell lines with IL-6 and FN-adhesion may confer a greater protection against chemotherapeutics than either effector alone. However, MTT cytotoxicity assays demonstrate that although adhesion to FN provides significant protection against treatment with mitoxantrone or doxorubicin (p=0.0002 and p=&lt;0.0001 respectively), the addition of IL-6 provides no further protection. These findings were corroborated by analysis of drug-mediated apoptosis using FCM by Annexin-V/7-AAD. In regards to cell cycle kinetics, our laboratory has previously demonstrated that adhesion of the 8226 MM cell line to FN mediated a p27Kip1 dependent G0/G1 cell cycle arrest. As predicted, BrdU/PI analysis of 8226 cells adhered to FN for 24 hours results in an increased number of cells in G0/G1 relative to cells maintained in suspension (p=0.0028). In contrast, when cells were adhered to FN in the presence of IL-6 no accumulation of cells in G0/G1 was observed, with levels similar to that observed in cells maintained in suspension with or without stimulation by IL-6. Our studies demonstrated that the G1/S cell cycle arrest associated with FN adhesion of MM cell lines was overcome when IL-6 was added; however, the cell adhesion mediated drug resistance (CAM-DR) was maintained in the presence of IL-6. Investigation of the biochemical signaling following concomitant exposure of MM cells to IL-6 and FN adhesion revealed a synergistic increase in STAT3 phosphorylation, nuclear translocation and DNA-binding as compared to either IL-6 or FN-adhesion alone in four MM cell lines. STAT3 phosphorylation was increased in cells adhered to FN in an IL-6 dose dependent manner. Electrophoretic mobility shift assay demonstrated a parallel 3-fold increase in STAT3/DNA complexes in cells adhered to FN relative to cells in suspension. To further characterize the receptor proximal affects of FN adhesion on IL-6 signaling we immunoprecipitated the IL-6R complex with antisera to gp130. Immunoprecipitation of gp130 revealed enhanced tyrosine phosphorylation of the gp130/Jak family complexes following stimulation FN-adhered RPMI 8226 MM cells with IL-6. Consistent with increased phosphorylation of the receptor complex, increased levels of phospho-STAT3 were identified associated with gp130 under co-stimulatory conditions relative to IL-6 or FN adhesion alone. Interestingly, immunoprecipitation with gp130 antibodies also revealed an association between STAT3 (non-phosphorylated) and gp130 in the absence of IL-6 stimulation in cells adhered to FN. These results suggest that adhesion to FN facilitates an IL-6-independent association between gp130 and STAT3, resulting in enhanced STAT3 signaling. Taken together, these data demonstrate a novel mechanism by which collaborative signaling by β1 integrin and gp130 confer an increased survival advantage to MM cells.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2456-2456
Author(s):  
Bin Lu ◽  
Xueqin Sun ◽  
John Crispino ◽  
Zan Huang

Abstract Genetic alterations as the initiator inducing leukemia also cause disorder of gene expression. Cooperation of these factors is known to be essential for leukemia development and remains to be further elucidated. In this study, we discovered that ANP32A dysregulation might contribute to myeloid leukemia. ANP32A expression was unanimously elevated in multiple online leukemia datasets and its upregulation was confirmed in human primary myeloid leukemia cells. Although ectopic expression of ANP32A did not promote leukemic cell proliferation, it did increase cell resistance to drug treatment such as TPA, Ara-C, VP16, and BCL2 inhibitor. Interestingly, ANP32A knockdown reduced cell proliferation and impaired colony formation in soft agar in various leukemic cells. ANP32A knockdown did so by inducing apoptosis and cell cycle arrest at G1 phase evidenced by upregulation of pro-apoptosis genes (BAK, BAD, cleaved caspase 3 and PARP) and downregulation of pro-survival or cell cycle progress genes (BCL2, CDK4, CCND1). The function of ANP32A knockdown to induce apoptosis and reduce colony formation was verified in human primary acute myeloid leukemia (AML) cells (Figure 1). However, re-introduction of BCL2, CDK4, or CCND1 failed to restore the impaired ability of colony formation by ANP32A knockdown, suggesting that apoptosis and cell cycle arrest may not be the direct effect of ANP32A knockdown. To probe how ANP32A would affect cell proliferation, we performed microarray analysis to identify potential ANP32A target genes including APOC1 and CCL26. Indeed, reintroduction of APOC1 or CCL26 significantly rescued colony formation ability while knockdown of APOC1 or CCL26 alone was sufficient to reduce colony formation. Further gene set enrichment analysis (GSEA) also revealed Notch signaling and histone modification signatures in ANP32A knockdown cells. To support this, ANP32A knockdown reduced intracellular Notch and NOTCH1 ovexpression significantly recovered the ability of colony formation. Furthermore, ANP32A knockdown also led to a global alteration of histone modifications including decrease of H3K9-acetylation (H3K9ac), H3K27-trimethylation (H3K27me3), and H3K4-trimethylation (H3K4me3) and increase of H3K9-trimethylation (H3K9me3). In fact, ChIP-PCR demonstrated that ANP32A bound to the promoter region of multiple target genes identified in microarray analysis and ANP32A downregulation caused decreased H3K27me3 and H3K4me3 at the same sites (Figure 2). These findings suggest that ANP32A may bind to the chromosome and involve histone modifications. To further test the potential function of ANP32A in leukemogenesis, we took advantage of MLL-AF9 fusion gene to immortalize bone marrow cells in vitro and compared wild-type (WT) cells to Anp32a knockout (Anp32a-/-) cells. We found that MLL-AF9-transduced Anp32a-/- bone marrow cells reproduced all phenotypes of leukemic cells with ANP32A knockdown: Anp32a-/- cells proliferated less, formed less colonies, exhibited more apoptosis, and showed cell cycle arrest at G1 phase compared to WT cells (Figure 3). These observations suggest a potential role of ANP32A in MLL-AF9-induced acute myeloid leukemia. Taken together, our studies have revealed ANP32A as a potential novel player to cooperate with other genetic alterations to induce leukemia. ANP32A upregulation may somehow involve histone modifications at genomic level that ultimately alter the expression of multiple genes and facilitate leukemic cell proliferation and survival. Thus ANP32A may serve as a potential target for developing novel therapy or biomarker for diagnosis and prognosis. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document