Graviola inhibits hypoxia-induced NADPH oxidase activity in prostate cancer cells reducing their proliferation and clonogenicity

Gagan Deep, Rahul Kumar, Anil K Jain, Deepanshi Dhar, Gati K Panigrahi, Anowar Hussain, Chapla Agarwal, Tamam El-Elimat, Vincent P Sica, Nicholas H Oberlies, Rajesh Agarwal, Gagan Deep, Rahul Kumar, Anil K Jain, Deepanshi Dhar, Gati K Panigrahi, Anowar Hussain, Chapla Agarwal, Tamam El-Elimat, Vincent P Sica, Nicholas H Oberlies, Rajesh Agarwal

Abstract

Prostate cancer (PCa) is the leading malignancy among men. Importantly, this disease is mostly diagnosed at early stages offering a unique chemoprevention opportunity. Therefore, there is an urgent need to identify and target signaling molecules with higher expression/activity in prostate tumors and play critical role in PCa growth and progression. Here we report that NADPH oxidase (NOX) expression is directly associated with PCa progression in TRAMP mice, suggesting NOX as a potential chemoprevention target in controlling PCa. Accordingly, we assessed whether NOX activity in PCa cells could be inhibited by Graviola pulp extract (GPE) that contains unique acetogenins with strong anti-cancer effects. GPE (1-5 μg/ml) treatment strongly inhibited the hypoxia-induced NOX activity in PCa cells (LNCaP, 22Rv1 and PC3) associated with a decrease in the expression of NOX catalytic and regulatory sub-units (NOX1, NOX2 and p47(phox)). Furthermore, GPE-mediated NOX inhibition was associated with a strong decrease in nuclear HIF-1α levels as well as reduction in the proliferative and clonogenic potential of PCa cells. More importantly, GPE treatment neither inhibited NOX activity nor showed any cytotoxicity against non-neoplastic prostate epithelial PWR-1E cells. Overall, these results suggest that GPE could be useful in the prevention of PCa progression via inhibiting NOX activity.

Figures

Figure 1. NOX expression is associated with…
Figure 1. NOX expression is associated with disease progression in TRAMP mice.
(A,B) NOX1 and p67phox expression was analyzed by IHC in TRAMP prostate tissue with different stages of the disease as well as in non-transgenic mice prostate tissue as negative control. Representative photographs are presented at 100x; inset represent further magnification of a part of the photograph. Immunoreactivity (represented by brown staining) of NOX1 and p67phox was scored as described in methods. Abbreviations: NC: Negative control; LG: Low-grade; HG: High-grade; PIN: Prostate intra-epithelial neoplasia; WD: Well differentiated; MD: Moderately differentiated; PD: Poorly differentiated. *p ≤ 0.001.
Figure 2. Comparison of various parts of…
Figure 2. Comparison of various parts of Graviola for cytotoxicity against PCa cells.
(A,B) LNCaP and PC3 cells were seeded at the density of 5 × 104 cells/well in 6-well plates. After 24 h of seeding, cells were treated with 25 and 50 μg/ml concentrations of Graviola extracts (D1: seeds extract; D2: pulp extract; D3: exocarp extract; D4: leaves extract; D5: twigs extract) for 48 h. At the end of 48 h, cells were harvested and counted as mentioned in ‘Materials and Methods’, and total cell number and percentage of dead cells are presented as mean ± SEM. *p ≤ 0.001; #p ≤ 0.01.
Figure 3. GPE inhibits NOX activity in…
Figure 3. GPE inhibits NOX activity in human PCa cells.
(A–C) Human PCa 22Rv1, LNCaP and PC3 cells were seeded at the density of 4 × 105 cells/60 mm culture dishes. After 24 h of seeding, cells were treated with 1, 2.5 and 5 μg/ml concentrations of GPE under hypoxic condition (1% O2) for 24 h. In each case, cells cultured under normoxic condition (21% O2) served as relevant control. At the end of 24 h, cells were harvested and NOX activity was measured as mentioned in ‘Materials and Methods’ and represented as rlu/mg protein. (D) Direct inhibition of NOX activity by GPE was assessed by pre-incubating GPE (1.0–5.0 μg/ml) with cellular homogenates and NOX activity was measured as mentioned in ‘Materials and Methods’ and represented as rlu/mg protein. Each value represents mean ± SEM of three samples for each treatment. $p ≤ 0.05; #p ≤ 0.01; *p ≤ 0.001.
Figure 4. GPE inhibits the expression of…
Figure 4. GPE inhibits the expression of members of NOX system and HIF-1α in human PCa 22Rv1 cells.
(A) 22Rv1 cells were treated with GPE (1–10 μg/ml) under normoxic (21% O2) or hypoxic (1% O2) conditions for 24 h. Thereafter, cells were collected and analyzed for mRNA expression of NOX1, p47phox and HIF-1α by RT-PCR using gene specific primers as mentioned in ‘Materials and Methods’. β-actin served as an endogenous internal standard. (B) 22Rv1 cells were treated with GPE (1–5 μg/ml) under hypoxic (1% O2) condition for 24 h. Thereafter, cells were collected and cytosolic and membrane fractions were prepared and analyzed for levels of NOX1, NOX2, p67phox and p47phox by immunoblotting. β-actin and E-cadherin were used to check the purity of cytoplasmic and membrane fractions as well as for loading control. Densitometry data presented below the bands are ‘fold change’ as compared with control (DMSO treated) after normalization with respective loading control. (C) 22Rv1 cells were treated with DPI (20 μM) and GPE (10 and 20 μg/ml) under hypoxic (1% O2) condition for 6 h. Cells cultured under normoxic condition (21% O2) served as relevant control. Thereafter, cells were collected and total cell lysates were prepared and analyzed for HIF-1α, HIF-1β, HIF-2α and tubulin. Densitometry data presented below the bands are ‘fold change’ as compared with hypoxia control (DMSO treated) after normalization with respective loading control. (D) 22Rv1 cells were treated with GPE (10 and 20 μg/ml) under hypoxic (1% O2) condition for 6 h. Cells cultured under normoxic condition (21% O2) served as relevant control. Thereafter, cells were collected and nuclear/cytoplasmic fractions were prepared and analyzed for HIF-1α expression. Membranes were also probed for TBP and tubulin as loading control for nuclear and membrane fractions, respectively. Densitometry data presented below the bands are ‘fold change’ as compared with hypoxia control (DMSO treated) after normalization with respective loading control. Abbreviations: N: Normoxic; H: Hypoxic; GPE: Graviola pulp extract; DPI: Diphenyleneiodonium.
Figure 5. GPE inhibits the clonogenicity of…
Figure 5. GPE inhibits the clonogenicity of human PCa cells under normoxic and hypoxic conditions.
(A,B) Human PCa 22Rv1 and PC3 cells (~1 × 103 cells per well) were cultured in 6-well plates and treated with GPE (1–5 μg/ml) under normoxic (21% O2) or hypoxic (1% O2) conditions for 48 hrs. Thereafter, cells were maintained under normoxic conditions and clones were counted after 7 days. Each value represents mean ± SEM of three samples for each treatment. #p ≤ 0.01; *p ≤ 0.001.
Figure 6. GPE reduces cell viability of…
Figure 6. GPE reduces cell viability of human PCa cells under normoxic and hypoxic conditions.
(A,B) 22Rv1, LNCaP, PC3 and PWR-1E cells were seeded at the density of 5 × 104 cells/well in six well plates. After 24 h of seeding, cells were treated with 2.5–20 μg/ml concentrations of GPE under normoxic (21% O2) or hypoxic (1% O2) conditions for 24 and 48 h. At the end of each time point, cells were harvested and counted as mentioned in ‘Materials and Methods’, and total cell number and percentage of dead cells are shown. Each value represents mean ± SEM of three samples for each treatment. $p ≤ 0.05; #p ≤ 0.01; *p ≤ 0.001.
Figure 7. Chemical characterization of GPE.
Figure 7. Chemical characterization of GPE.
Sample was analyzed as detailed in the methods. (A) The total ion chromatogram of the GPE. (B) The extracted ion chromatograms, detailed to the right, overlaid with each other. The color coding subdivides the acetogenins based upon their molecular ions.
Figure 8. Example characterization of acetogenins.
Figure 8. Example characterization of acetogenins.
The MS/MS spectrum of the peak at 4.58 min (coded blue in Fig. 7). These data illustrate the common fragmentation patterns observed with acetogenins. In particular, note the fragments adjacent to hydroxy moieties, thereby facilitating the elucidation of the positioning of the THF ring and each hydroxy group. Based on these data and the associated literature, this compound is tentatively identified as annonacin.

References

    1. Siegel R. L., Miller K. D. & Jemal A. Cancer statistics, 2015. CA Cancer J Clin 65, 5–29 (2015).
    1. Kumar B., Koul S., Khandrika L., Meacham R. B. & Koul H. K. Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res 68, 1777–1785 (2008).
    1. Khandrika L., Kumar B., Koul S., Maroni P. & Koul H. K. Oxidative stress in prostate cancer. Cancer Lett 282, 125–136 (2009).
    1. Lim S. D. et al. Increased Nox1 and hydrogen peroxide in prostate cancer. Prostate 62, 200–207 (2005).
    1. Arbiser J. L. et al. Reactive oxygen generated by Nox1 triggers the angiogenic switch. Proc Natl Acad Sci USA 99, 715–720 (2002).
    1. Tam N. N., Gao Y., Leung Y. K. & Ho S. M. Androgenic regulation of oxidative stress in the rat prostate: involvement of NAD(P)H oxidases and antioxidant defense machinery during prostatic involution and regrowth. Am J Pathol 163, 2513–2522 (2003).
    1. Block K. & Gorin Y. Aiding and abetting roles of NOX oxidases in cellular transformation. Nat Rev Cancer 12, 627–637 (2012).
    1. Li Q. et al. NADPH oxidase subunit p22(phox)-mediated reactive oxygen species contribute to angiogenesis and tumor growth through AKT and ERK1/2 signaling pathways in prostate cancer. Biochim Biophys Acta 1833, 3375–3385 (2013).
    1. Lambeth J. D. Nox enzymes, ROS, and chronic disease: an example of antagonistic pleiotropy. Free Radic Biol Med 43, 332–347 (2007).
    1. Kamata T. Roles of Nox1 and other Nox isoforms in cancer development. Cancer Sci 100, 1382–1388 (2009).
    1. Suh Y. A. et al. Cell transformation by the superoxide-generating oxidase Mox1. Nature 401, 79–82 (1999).
    1. Itoh T. et al. Cisplatin induces production of reactive oxygen species via NADPH oxidase activation in human prostate cancer cells. Free Radic Res 45, 1033–1039 (2011).
    1. Suzuki S. et al. Apocynin, an NADPH oxidase inhibitor, suppresses rat prostate carcinogenesis. Cancer Sci 104, 1711–1717 (2013).
    1. Antony S. et al. Characterization of NADPH oxidase 5 expression in human tumors and tumor cell lines with a novel mouse monoclonal antibody. Free Radic Biol Med 65, 497–508 (2013).
    1. Suzuki S., Pitchakarn P., Sato S., Shirai T. & Takahashi S. Apocynin, an NADPH oxidase inhibitor, suppresses progression of prostate cancer via Rac1 dephosphorylation. Exp Toxicol Pathol 65, 1035–1041 (2013).
    1. Diebold I., Petry A., Hess J. & Gorlach A. The NADPH oxidase subunit NOX4 is a new target gene of the hypoxia-inducible factor-1. Mol Biol Cell 21, 2087–2096 (2010).
    1. Yuan G. et al. Hypoxia-inducible factor 1 mediates increased expression of NADPH oxidase-2 in response to intermittent hypoxia. J Cell Physiol 226, 2925–2933 (2011).
    1. Block K. et al. NAD(P)H oxidases regulate HIF-2alpha protein expression. J Biol Chem 282, 8019–8026 (2007).
    1. Ushio-Fukai M. & Nakamura Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett 266, 37–52 (2008).
    1. Ramteke A. et al. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol Carcinog 54, 554–565 (2015).
    1. Butterworth K. T. et al. Hypoxia selects for androgen independent LNCaP cells with a more malignant geno- and phenotype. Int J Cancer 123, 760–768 (2008).
    1. Ranasinghe W. K. et al. The role of hypoxia-inducible factor 1alpha in determining the properties of castrate-resistant prostate cancers. Plos One 8, e54251 (2013).
    1. Dai Y., Bae K. & Siemann D. W. Impact of hypoxia on the metastatic potential of human prostate cancer cells. Int J Radiat Oncol Biol Phys 81, 521–528 (2011).
    1. Morton J. Soursop In Fruits of warm climates (ed Morton J.) 75–80 (Echo Point Books and Media, 1987).
    1. Dai Y. et al. Selective growth inhibition of human breast cancer cells by graviola fruit extract in vitro and in vivo involving downregulation of EGFR expression. Nutr Cancer 63, 795–801 (2011).
    1. Torres M. P. et al. Graviola: a novel promising natural-derived drug that inhibits tumorigenicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering cell metabolism. Cancer Lett 323, 29–40 (2012).
    1. Raina K. et al. Stage-specific inhibitory effects and associated mechanisms of silibinin on tumor progression and metastasis in transgenic adenocarcinoma of the mouse prostate model. Cancer Res 68, 6822–6830 (2008).
    1. Raina K. et al. Dietary feeding of silibinin inhibits prostate tumor growth and progression in transgenic adenocarcinoma of the mouse prostate model. Cancer Res 67, 11083–11091 (2007).
    1. Sun S., Liu J., Kadouh H., Sun X. & Zhou K. Three new anti-proliferative Annonaceous acetogenins with mono-tetrahydrofuran ring from graviola fruit (Annona muricata). Bioorg Med Chem Lett 24, 2773–2776 (2014).
    1. Brar S. S. et al. NOX5 NAD(P)H oxidase regulates growth and apoptosis in DU 145 prostate cancer cells. Am J Physiol Cell Physiol 285, C353–369 (2003).
    1. Le Ven J. et al. Comprehensive characterization of Annonaceous acetogenins within a complex extract by HPLC-ESI-LTQ-Orbitrap(R) using post-column lithium infusion. J Mass Spectrom 47, 1500–1509 (2012).
    1. Fall D., Duval R. A., Gleye C., Laurens A. & Hocquemiller R. Chamuvarinin, an acetogenin bearing a tetrahydropyran ring from the roots of Uvaria chamae. J Nat Prod 67, 1041–1043 (2004).
    1. Allegrand J. et al. Structural study of acetogenins by tandem mass spectrometry under high and low collision energy. Rapid Commun Mass Spectrom 24, 3602–3608 (2010).
    1. Maraldi T. Natural compounds as modulators of NADPH oxidases. Oxid Med Cell Longev 2013, 271602 (2013).
    1. Jaquet V., Scapozza L., Clark R. A., Krause K. H. & Lambeth J. D. Small-molecule NOX inhibitors: ROS-generating NADPH oxidases as therapeutic targets. Antioxid Redox Signal 11, 2535–2552 (2009).
    1. McLaughlin J. L. Paw paw and cancer: annonaceous acetogenins from discovery to commercial products. J Nat Prod 71, 1311–1321 (2008).
    1. Oberlies N. H., Croy V. L., Harrison M. L. & McLaughlin J. L. The Annonaceous acetogenin bullatacin is cytotoxic against multidrug-resistant human mammary adenocarcinoma cells. Cancer Lett 115, 73–79 (1997).
    1. Morre D. J., de Cabo R., Farley C., Oberlies N. H. & McLaughlin J. L. Mode of action of bullatacin, a potent antitumor acetogenin: inhibition of NADH oxidase activity of HeLa and HL-60, but not liver, plasma membranes. Life Sci 56, 343–348 (1995).
    1. Alali F. Q., Liu X. X. & McLaughlin J. L. Annonaceous acetogenins: recent progress. J Nat Prod 62, 504–540 (1999).
    1. El-Elimat T. et al. Greensporones: resorcylic acid lactones from an aquatic Halenospora sp. J Nat Prod 77, 2088–2098 (2014).
    1. Schlaepfer I. R. et al. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget 6, 22836–22856 (2015).
    1. Deep G., Inturi S. & Agarwal R. Methods to analyze chemopreventive effect of silibinin on prostate cancer biomarkers protein expression In Cancer Prevention: Dietary Factors and Pharmacology (eds Bode A. & Dong Z.) 85–105 (Humana Press, 2013).

Source: PubMed

Подписаться