International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Bladder cancer (BC) is one of the most prevalent malignancies globally, with an estimated 613,791 new cases reported in 2022 according to GLOBOCAN 2022(1). Clinically, BC is categorized into three main subtypes, namely non-muscle-invasive (NMI), MIB and metastatic BC (2). While NMIBC accounts for ~80% of initial BC diagnoses and is associated with a favorable 5-year survival rate >90% (3), it has a 1-year recurrence rate of 15-61%, and a 5-year recurrence rate of 31-78% (4). Its high recurrence rate imposes substantial healthcare burdens (5-9). By contrast, MIBC and metastatic BC, accounting for ~25% of all diagnoses, are associated with aggressive progression and poorer prognosis (10). Disparities in BC outcomes are associated with ethnicity, sex and socioeconomic factors (11-16). Management strategies for NMIBC and MIBC include transurethral resection combined with intravesical therapies, such as mitomycin C and Bacillus Calmette-Guérin (17-21), and platinum-based neoadjuvant chemotherapy followed by radical cystectomy, respectively (10,22). However, chemotherapy resistance and toxicity underscore the need for more safe and effective therapeutic agents (22).
Pueraria spp. (Leguminosae), notably P. lobata and P. thomsonii, are medicinal plants that are widely distributed in East Asia. The 2020 edition of the Chinese Pharmacopoeia recognizes the dried roots (radix) of Pueraria as a source of bioactive compounds, particularly isoflavonoids such as puerarin, which are the primary active components of Puerariae radix flavones (PRF) (23-43). Previous studies demonstrated that PRF exhibit multifaceted pharmacological properties, such as anti-cancer effects, in colon (HT-29 cells), cervical (HeLa cells), liver (SMMC-7721 cells) and breast cancer (HS578T, MDA-MB-231 and MCF-7 cells), acute promyelocytic leukemia (NB4 cells), neuroblastoma (SHSY5Y cells), pancreatic carcinoma (BxPC-3 cells) and lung cancer (A549 cells) (44-48). Despite these findings, the anti-tumor activity of PRF against BC remains unexplored.
T24 cells serve as the preferred model for research on invasion, metastasis and drug resistance mechanisms in BC. Their intrinsic cisplatin resistance (49) makes T24 cells a classical system for exploring drug mechanisms of action (50) that are widely used in fundamental BC research (51,52). Moreover, in natural compound anticancer mechanism studies, most researchers employ single-cell models to investigate mechanisms of action (47,50). Therefore, the present study investigates the effects of PRF on human bladder cancer T24 cells and its underlying molecular mechanisms.
PRF (Taobao; purity ≥80%) was dissolved in anhydrous ethanol to prepare an 8,000 µg/ml stock solution, which was stored at 4˚C. T24 cells (Procell Life Science & Technology Co., Ltd.) were authenticated by STR profiling (Mayo Clinic Cytogenetics Core) and confirmed to be free of mycoplasma contamination. Cells were cultured in RPMI-1640 (Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Shanghai VivaCell Biosciences, Ltd.) at 37˚C in a humified atmosphere with 5% CO2. Cells were maintained at a density of <1x106 cells/ml and used within three passages.
Cells (5x10³/well) in 200 µl medium were treated with 0, 25, 50, 100 and 200 µg/ml PRF at 37˚C for 24 or 48 h. Following treatment with MTT reagent (Sigma-Aldrich, Merck KGaA), cells were incubated at 37˚C for an additional 4 h. Finally, the absorbance at 490 nm was measured using a microplate reader.
Following treatment with 0-200 µg/ml PRF at 37˚C for 24 or 48 h, T24 cells were stained with AO/EB (Sigma-Aldrich, Merck KGaA) in the dark at room temperature for 5 min. Images were captured under a fluorescence microscope (magnification, x200).
Following incubation with PRF (0, 50, 100 and 200 µg/ml) at 37˚C for 24 or 48 h, 1x106 T24 cells were collected. Subsequently, genomic DNA was extracted from untreated and PRF-treated cells using the Apoptosis DNA Ladder Extraction kit (cat. no. #C0007; Beyotime Institute of Biotechnology) according to the manufacturer's instructions. Finally, 5 µl DNA (containing 1 µg DNA) was mixed with 6X Loading Buffer and loaded onto a 1% agarose gel prepared in TBE buffer containing 0.5 µg/ml ethidium bromide (EB; Thermo Fisher Scientific, Inc.). Electrophoresis was performed at a constant voltage of 100 V for 30 min. DNA bands were visualized under UV irradiation (302 nm) using a gel imaging system. Caution: Ethidium bromide is a mutagenic agent. All procedures were conducted with nitrile gloves, and waste was disposed as hazardous material.
Following the manufacturer's protocols, total RNA was extracted from cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), and reverse-transcribed into cDNA using the Transcriptor First Strand cDNA synthesis kit (cat. no. #4897030001; Roche Diagnostics). cDNA amplification was performed in triplicate with the FastStart Universal SYBR Green Master (ROX) kit (cat. no. #4913850001; Roche Diagnostics) on an ABI Prism 7500 system (Thermo Fisher Scientific, Inc.). The primer sequences for BCL2, BAX, FAS receptor (FAS), FAS ligand (FASL), tumor necrosis factor receptor 1 (TNFR1), tumor necrosis factor-α (TNF-α), cysteinyl aspartate-specific protease-3 (CASP3), NF-κB and GAPDH are listed in Table I. The primers were designed and synthesized by Sangon Biotech Co., Ltd.
The thermocycling conditions were as follows: Initial denaturation at 50˚C for 2 min and 95˚C for 10 min, followed by 45 cycles of 95˚C for 15 sec and 60˚C for 1 min. The relative mRNA expression levels were calculated using the 2-ΔΔCq method (53).
Following incubation with PRF (0, 50, 100 and 200 µg/ml) at 37˚C for 24 or 48 h, cells and culture medium were collected. The protein expression levels of FAS and TNF-α were quantified in T24 cells and culture supernatant using the corresponding ELISA kits (cat. nos. #JL14207 and #JL10208, respectively; both Shanghai Future Industrial Co., Ltd.) according to the manufacturer's instructions.
All data were analyzed using SPSS Statistics (version 25; IBM Corporation) or GraphPad Prism (version 8.1; GraphPad Software, Inc.; Dotmatics). Experiments were performed in triplicate, and all data are presented as the mean ± SD. Data were analyzed using two-way repeated-measures ANOVA followed by Dunnett's post hoc tests. P<0.05 was considered to indicate a statistically significant difference.
MTT assay demonstrated that PRF could significantly inhibit the viability of T24 cells in a time- and dose-dependent manner (Fig. 1A). The inhibition rate in cells treated with 200 µg/ml PRF for 12 h was significantly higher compared with that in the control group (Fig. 1A). Additionally, the inhibition rates of cells exposed to 50, 100 and 200 µg/ml PRF for 24 h were all increased compared with the control group. Consistently, at 48 and 72 h, the inhibition rates in the 100 and 200 µg/ml PRF-treated groups were significantly higher compared with the control group. PRF exhibited a clear dose- and time-dependent inhibitory effect on cell viability, with inhibition increasing with enhanced concentration and exposure duration (Fig. 1B). Significant differences in inhibition rates were observed between time points (12, 24, 48 and 72 h).
AO/EB staining revealed distinct apoptotic progression in T24 cells (Fig. 2). Increasing PRF concentrations induced a progressive fluorescence shift from green to orange-red emission. Morphologically, cells progressed from intact plasma membranes with uniform chromatin distribution to display early apoptotic hallmarks (cellular shrinkage, nuclear condensation, and apoptotic body formation) and late-stage apoptosis typified by membrane rupture, chromatin fragmentation, and nuclear dissolution. Control cells exhibited a uniform green staining (Fig. 2A and E). However, 50 µg/ml PRF induced early apoptotic features, such cell shrinkage, condensed nuclei and apoptotic bodies (Fig. 2B and F). Additionally, higher PRF concentrations (100-200 µg/ml) induced features of late apoptosis, including enhanced membrane permeability, chromatin fragmentation (orange/red staining) and nuclear disintegration (Fig. 2C, D, G and H). Notably, 100-200 µg/ml PRF exposure for 24-48 h promoted concentration- and time-dependent morphological alterations, characteristic of late-stage apoptosis.
The DNA ladder assay showed no apoptotic fragmentation in T24 cells treated with PRF (0-200 µg/ml) for 24-48 h, thus suggesting that PRF-induced apoptosis was triggered via a DNA fragmentation-independent pathway (Fig. 3).
RT-qPCR demonstrated that treatment with 100 or 200 µg/ml PRF for 24 h upregulated FAS (2.0- and 2.3-fold, respectively), FASL (48.9- and 15.2-fold, respectively), TNF-α (1.4- and 2.2-fold, respectively) and CASP3 (2.1- and 1.8-fold, respectively; Fig. 4A-C and E). However, the expression of TNFR1 only significantly increased in cells treated with 200 µg/ml PRF (1.2-fold; Fig. 4D). No significant changes were observed in the mRNA expression of NF-κB, BCL-2 and BAX (Fig. 4F-H). Additionally, treatment with PRF for 48 h slightly upregulated FAS at 100 and 200 µg/ml (1.3- and 1.2-fold, respectively; Fig. 4A), and markedly upregulated TNF-α at 200 µg/ml (1.6-fold; Fig. 4C). By contrast, FASL (0.5-fold at 100 µg/ml), TNFR1 (0.8-fold at 200 µg/ml), and NF-κB (0.8- and 0.6-fold at 100 and 200 µg/ml, respectively) were significantly downregulated (Fig. 4B, D and F). No significant alterations were detected in the mRNA expression of CASP3, BCL-2 and BAX (Fig. 4E, G and H).
ELISA showed that treatment with PRF for 24 h resulted in a dose-dependent increase of intracellular FAS and TNF-a protein levels (50-200 µg/ml; Fig. 5A and C). By contrast, the secreted levels of FAS were reduced in the culture supernatant (50-200 µg/ml; Fig. 5B). In addition, the secreted levels of TNF-α were elevated only at 50 µg/ml (Fig. 5D). Furthermore, T24 cell exposure to PRF for 48 h dose-dependently increased the intracellular and secreted levels of FAS (50-200 µg/ml; Fig. 5A and B). However, intracellular and secreted TNF-α levels were only significantly elevated at 50 µg/ml (Fig. 5C and D).
The present study demonstrated that treatment with PRF for 24 and 48 h significantly inhibited T24 cell viability in a dose- and time-dependent manner, with the most pronounced effects observed at 100 and 200 µg/ml. The ability of PRF to promote T24 cell apoptosis was shown by flow cytometry in our previous study (54). Here, morphological examination revealed characteristic apoptotic features, including early-stage membrane shrinkage and late-stage apoptotic body formation. Notably, the absence of DNA laddering suggested that PRF induced apoptosis via a non-canonical DNA fragmentation pathway. Although no direct evidence currently demonstrates PRF-induced apoptosis via this mechanism in other cancer cells, it is hypothesized that negative DNA fragmentation results may contribute to the underreporting of such findings. Consistent with this observation, our team previously demonstrated flavonoid compounds extracted from Galium verum L. likewise yield negative DNA fragmentation results when inducing apoptosis in HepG2 hepatocellular carcinoma cells (unpublished data). The hypothesis that PRF induces apoptosis through non-classical DNA fragmentation pathways is based on the T24 cell model. To the best of our knowledge, the present study is the first to report a non-canonical DNA fragmentation route during PRF-induced apoptosis in T24 cells.
PRF treatment significantly upregulated FAS, FASL, TNFR1, TNFα and CASP3 in T24 cells, while NF-κB was downregulated. mRNA expression levels of both BCL2 and BAX remained unchanged, indicating that PRF-mediated apoptosis in T24 cells may involve the coordinated regulation of FAS, FASL, TNFR1, TNFα, CASP3 and NF-κB expression dynamics (Fig. 6).
Binding of FASL to its receptor induces receptor trimerization and activation (55,56). Subsequently, activated FAS can recruit Fas-associated death domain (FADD), which undergoes conformational changes that allow it to bind and cleave procaspase-8, promoting the formation of the death-inducing signaling complex (DISC). Within this complex, activated caspase-8 propagates the apoptotic cascade via cleaving and activating caspase-3 (55,56). Here, the pronounced upregulation of FAS, FASL and CASP3, accompanied by the increased intracellular FAS protein levels, supported the activation of the extrinsic apoptotic pathway. Protein expression levels of FAS were markedly decreased in cell supernatant following treatment with PRF for 24 h, and significantly elevated at 48 h. During early apoptosis, FAS proteins may aggregate and bind to the plasma membrane, leading to elevated intracellular FAS levels. In late apoptosis, loss of plasma membrane integrity may facilitate the release of membrane-bound FAS proteins into the culture medium in a soluble form (57-59), resulting in the observed increase in soluble FAS protein levels at 48 h.
DISC-associated caspase-8 can undergo auto-proteolysis to activate caspase-8, which cleaves interacting domain death agonist (Bid) into truncated (t)Bid (56,60). tBid can translocate to mitochondria, thus perturbing the equilibrium between pro-apoptotic and anti-apoptotic Bcl-2 family members (60-62). However, the lack of alterations in the mRNA expression levels of BCL2 and BAX indicated that the PRF-induced cell apoptosis may occur independently of the mitochondrial pathway. Nandana et al (50) confirmed that Brucein D (a bioactive quassinoid compound isolated from Brucea javanica fruit) induces apoptosis in T24 cells by regulating the Bcl-2/Bax pathway, exhibiting the classical DNA ladder feature. Conversely, the typical DNA laddering would not be expected in apoptotic mechanisms independent of the Bcl-2/Bax pathway. This was further reinforced by the absence of characteristic DNA ladder fragmentation, supporting extrinsic apoptotic pathway involvement.
In parallel with the FAS pathway, TNFα binding to TNFR1 disrupts its interaction with the inhibitory protein silencer of death domains, enabling the recruitment of TNF receptor-associated death domain (TRADD) through the intracellular death domain (63-67). TRADD interacts with FADD, activating procaspase-8 to caspase-8, which cleaves caspase-3. In the present study, the concurrent upregulation of TNFR1, TNFα and CASP3 further supported the involvement of this TNFα-driven apoptotic mechanism. In addition, the significant elevation of the TNF-α protein expression demonstrated that TNF-α may be involved in PRF-induced apoptosis in BC cells.
TRADD recruits TNFR-associated factor 2 and receptor-interacting serine/threonine-protein kinase 1, leading to the activation of the IKK complex, which degrades IκB (63,64). This process can promote the release of NF-κB, allowing its nuclear translocation and the subsequent transcription of pro-survival genes (63,64). Paradoxically, the present NF-κB downregulation indicated that PRF may suppress this survival signaling axis, thus shifting the cellular balance toward apoptosis. Although this interaction may be indirect, the inverse association between NF-κB suppression and caspase activation may provide evidence for the pro-apoptotic effects of PRF.
NF-κB exhibits biphasic regulation (68). NF-κB upregulation promotes cellular survival via transcription of pro-survival genes (63,64). However, its pathological hyperactivation promotes tumor progression and chemoresistance through the upregulation of anti-apoptotic factors and activation of survival signaling (69,70). By contrast, crosstalk between TNF-α and FAS could promote the establishment of a compensatory apoptotic axis by amplifying the activation of caspase-8 and -3, thus counteracting NF-κB-mediated cytoprotection (71). In the present study, PRF led to a biphasic modulation of NF-κB, which was characterized by transient upregulation at 24 h followed by downregulation at 48 h. The aforementioned finding was consistent with context-dependent biphasic NF-κB regulation, implicating NF-κB in PRF-induced apoptosis. These results indicated a dual role for NF-κB in T24 cells, acting as both a pro-survival mediator and a stress-responsive modulator of apoptosis.
The present study had limitations. Firstly, the antitumor effects of PRF were not validated through in vivo animal experiments. Secondly, the findings were verified solely in the T24 cell line without replication in other BC cell lines, thereby restricting the generalizability of the conclusions. Investigation of the mechanistic pathways lacked protein-level validation by western blot analysis. Fourthly, no positive control was included in the DNA ladder assay.
In conclusion, the present study demonstrated that PRF exhibited a significant inhibitory effect on the viability of BC cells (T24 cell line). This inhibitory activity displayed a time- and dose-dependent association, with more pronounced effects observed at PRF concentrations of 100 and 200 µg/ml following treatment for 24 and 48 h. Mechanistically, the results indicated that the PRF-induced apoptosis was triggered by two mechanisms, namely the death receptor-mediated pathways (FAS/TNFR1) in the absence of mitochondrial pathways and the biphasic modulation of NF-κB signaling, characterized by a switch from survival to apoptosis. Overall, these findings highlighted the therapeutic potential of PRF for the treatment of apoptosis-resistant BC.
Not applicable.
Funding: The present study was supported by the National Natural Science Foundation of China (grant nos. 82173568, 81703269, 81973097 and 82273687), the Natural Science Foundation of Heilongjiang Province (grant no. LH2023H054), the Huoju Plan Research Foundation of Mudanjiang Medical University (grant no. 2022-MYHJ-014), the Special Program for Supervisor Scientific Research of Mudanjiang Medical University (grant nos. YJSZX2022137 and YJSZX2022141), the Administration of Science & Technology Foundation of Mudanjiang (grant nos. HT2022NS112 and HT2022NS113), the Scientific Research Project of the Health Commission of Heilongjiang Province (grant no. 20221212060609) and the Fundamental Research Funds for the Universities of Heilongjiang Province (grant no. 2022-KYYWFMY-0712).
The data generated in the present study may be requested from the corresponding author.
JD designed the study, analyzed and interpreted data and wrote and revised the manuscript. YG and HG designed the study and analyzed data. TJ and YM analyzed data and wrote the manuscript. SR designed the study and wrote and revised the manuscript. JD and SR confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Tran L, Xiao JF, Agarwal N, Duex JE and Theodorescu D: Advances in bladder cancer biology and therapy. Nat Rev Cancer. 21:104–121. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Katims AB, Tallman J, Vertosick E, Porwal S, Dalbagni G, Cha EK, Smith R, Benfante N and Herr HW: Response to 2 induction courses of bacillus Calmette-Guèrin therapy among patients with high-risk non-muscle-invasive bladder cancer: 5-Year follow-up of a phase 2 clinical trial. JAMA Oncol. 10:522–525. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Teoh JY, Kamat AM, Black PC, Grivas P, Shariat SF and Babjuk M: Recurrence mechanisms of non-muscle-invasive bladder cancer-a clinical perspective. Nat Rev Urol. 19:280–294. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Knowles MA and Hurst CD: Molecular biology of bladder cancer: New insights into pathogenesis and clinical diversity. Nat Rev Cancer. 15:25–41. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Babjuk M, Böhle A, Burger M, Capoun O, Cohen D, Compérat EM, Hernández V, Kaasinen E, Palou J, Rouprêt M, et al: EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: Update 2016. Eur Urol. 71:447–461. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Berdik C: Unlocking bladder cancer. Nature. 551:S34–S35. 2017.PubMed/NCBI View Article : Google Scholar | |
|
James AC and Gore JL: The costs of non-muscle invasive bladder cancer. Urol Clin North Am. 40:261–269. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Abdollah F, Gandaglia G, Thuret R, Schmitges J, Tian Z, Jeldres C, Passoni NM, Briganti A, Shariat SF, Perrotte P, et al: Incidence, survival and mortality rates of stage-specific bladder cancer in United States: A trend analysis. Cancer Epidemiol. 37:219–225. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Witjes JA, Bruins HM, Cathomas R, Compérat EM, Cowan NC, Gakis G, Hernández V, Linares Espinós E, Lorch A, Neuzillet Y, et al: European association of urology guidelines on muscle-invasive and metastatic bladder cancer: Summary of the 2020 guidelines. Eur Urol. 79:82–104. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Wang Y, Chang Q and Li Y: Racial differences in urinary bladder cancer in the United States. Sci Rep. 8(12521)2018.PubMed/NCBI View Article : Google Scholar | |
|
Schinkel JK, Shao S, Zahm SH, McGlynn KA, Shriver CD and Zhu K: Overall and recurrence-free survival among black and white bladder cancer patients in an equal-access health system. Cancer Epidemiol. 42:154–158. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Kaye DR, Canner JK, Kates M, Schoenberg MP and Bivalacqua TJ: Do African American patients treated with radical cystectomy for bladder cancer have worse overall survival? Accounting for pathologic staging and patient demographics beyond race makes a difference. Bladder Cancer. 2:225–234. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Casey MF, Gross T, Wisnivesky J, Stensland KD, Oh WK and Galsky MD: The impact of regionalization of cystectomy on racial disparities in bladder cancer care. J Urol. 194:36–41. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Dobruch J, Daneshmand S, Fisch M, Lotan Y, Noon AP, Resnick MJ, Shariat SF, Zlotta AR and Boorjian SA: Gender and bladder cancer: A collaborative review of etiology, biology, and outcomes. Eur Urol. 69:300–310. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Radkiewicz C, Edgren G, Johansson ALV, Jahnson S, Häggström C, Akre O, Lambe M and Dickman PW: Sex Differences in urothelial bladder cancer survival. Clin Genitourin Cancer. 18:26–34.e6. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Babjuk M, Burger M, Compérat EM, Gontero P, Mostafid AH, Palou J, van Rhijn BWG, Rouprêt M, Shariat SF, Sylvester R, et al: European association of urology guidelines on non-muscle-invasive bladder cancer (TaT1 and carcinoma in situ)-2019 update. Eur Urol. 76:639–657. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Messing EM, Tangen CM, Lerner SP, Sahasrabudhe DM, Koppie TM, Wood DP Jr, Mack PC, Svatek RS, Evans CP, Hafez KS, et al: Effect of intravesical instillation of gemcitabine vs saline immediately following resection of suspected low-grade non-muscle-invasive bladder cancer on tumor recurrence: SWOG S0337 randomized clinical trial. JAMA. 319:1880–1888. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Monro S, Colón KL, Yin H, Roque J III, Konda P, Gujar S, Thummel RP, Lilge L, Cameron CG and McFarland SA: Transition metal complexes and photodynamic therapy from a tumor-centered approach: Challenges, opportunities, and highlights from the development of TLD1433. Chem Rev. 119:797–828. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Sylvester RJ, van der Meijden APM and Lamm DL: Intravesical bacillus Calmette-Guerin reduces the risk of progression in patients with superficial bladder cancer: A meta-analysis of the published results of randomized clinical trials. J Urol. 168:1964–1970. 2002.PubMed/NCBI View Article : Google Scholar | |
|
Lamm DL, Blumenstein BA, Crissman JD, Montie JE, Gottesman JE, Lowe BA, Sarosdy MF, Bohl RD, Grossman HB, Beck TM, et al: Maintenance bacillus Calmette-Guerin immunotherapy for recurrent TA, T1 and carcinoma in situ transitional cell carcinoma of the bladder: A randomized southwest oncology group study. J Urol. 163:1124–1129. 2000.PubMed/NCBI | |
|
Hermans TJN, Voskuilen CS, van der Heijden MS, Schmitz-Dräger BJ, Kassouf W, Seiler R, Kamat AM, Grivas P, Kiltie AE, Black PC and van Rhijn BWG: Neoadjuvant treatment for muscle-invasive bladder cancer: The past, the present, and the future. Urol Oncol. 36:413–422. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Shi W, Yuan R, Chen X, Xin Q, Wang Y, Shang X, Cong W and Chen K: Puerarin reduces blood pressure in spontaneously hypertensive rats by targeting eNOS. Am J Chin Med. 47:19–38. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Shukla R, Pandey N, Banerjee S and Tripathi Y: Effect of extract of Pueraria tuberosa on expression of hypoxia inducible factor-1α and vascular endothelial growth factor in kidney of diabetic rats. Biomed Pharmacother. 93:276–285. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Satpathy S, Patra A, Ahirwar B and Hussain MD: Antioxidant and anticancer activities of green synthesized silver nanoparticles using aqueous extract of tubers of Pueraria tuberosa. Artif Cells Nanomed Biotechnol. 46 (Suppl 3):S71–S85. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Jearapong N, Chatuphonprasert W and Jarukamjorn K: Miroestrol, a phytoestrogen from Pueraria mirifica, improves the antioxidation state in the livers and uteri of β-naphthoflavone-treated mice. J Nat Med. 68:173–180. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Ahmad B, Khan S, Liu Y, Xue M, Nabi G, Kumar S, Alshwmi M and Qarluq AW: Molecular mechanisms of anticancer activities of Puerarin. Cancer Manag Res. 12:79–90. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Lin YJ, Hou YC, Lin CH, Hsu YA, Sheu JJ, Lai CH, Chen BH, Lee Chao PD, Wan L and Tsai FJ: Puerariae radix isoflavones and their metabolites inhibit growth and induce apoptosis in breast cancer cells. Biochem Biophys Res Commun. 378:683–688. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Zhang XL, Wang BB and Mo JS: Puerarin 6''-O-xyloside possesses significant antitumor activities on colon cancer through inducing apoptosis. Oncol Lett. 16:5557–5564. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Bulugonda RK, Kumar KA, Gangappa D, Beeda H, Philip GH, Muralidhara Rao D and Faisal SM: Mangiferin from Pueraria tuberosa reduces inflammation via inactivation of NLRP3 inflammasome. Sci Rep. 7(42683)2017.PubMed/NCBI View Article : Google Scholar | |
|
Pandey N, Yadav D, Pandey V and Tripathi VB: Anti-inflammatory effect of Pueraria tuberosa extracts through improvement in activity of red blood cell anti-oxidant enzymes. Ayu. 34:297–301. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Srivastava S, Pandey H, Singh SK and Tripathi YB: Anti-oxidant, anti-apoptotic, anti-hypoxic and anti-inflammatory conditions induced by PTY-2 against STZ-induced stress in islets. Biosci Trends. 13:382–393. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Shukla R, Banerjee S and Tripathi YB: Pueraria tuberosa extract inhibits iNOS and IL-6 through suppression of PKC-α and NF-kB pathway in diabetes-induced nephropathy. J Pharm Pharmacol. 70:1102–1112. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Jin SE, Son YK, Min BS, Jung HA and Choi JS: Anti-inflammatory and antioxidant activities of constituents isolated from Pueraria lobata roots. Arch Pharm Res. 35:823–837. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Kim JM, Lee YM, Lee GY, Jang DS, Bae KH and Kim JS: Constituents of the roots of Pueraria lobata inhibit formation of advanced glycation end products (AGEs). Arch Pharm Res. 29:821–825. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Sun Y, Zhang H, Cheng M, Cao S, Qiao M, Zhang B, Ding L and Qiu F: New hepatoprotective isoflavone glucosides from Pueraria lobata (Willd.) Ohwi. Nat Prod Res. 33:3485–3492. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Sook Kim Y, Soo Lee I and Sook Kim J: Protective effects of Puerariae radix extract and its single compounds on methylglyoxal-induced apoptosis in human retinal pigment epithelial cells. J Ethnopharmacol. 152:594–598. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Sucontphunt A, De-Eknamkul W, Nimmannit U, Dan Dimitrijevich S and Gracy RW: Protection of HT22 neuronal cells against glutamate toxicity mediated by the antioxidant activity of Pueraria candollei var. mirifica extracts. J Nat Med. 65:1–8. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Koirala P, Seong SH, Jung HA and Choi JS: Comparative evaluation of the antioxidant and anti-Alzheimer's disease potential of coumestrol and puerarol isolated from Pueraria lobata using molecular modeling studies. Molecules. 23(785)2018.PubMed/NCBI View Article : Google Scholar | |
|
Anukulthanakorn K, Parhar IS, Jaroenporn S, Kitahashi T, Watanbe G and Malaivijitnond S: Neurotherapeutic effects of Pueraria mirifica extract in early- and late-stage cognitive impaired rats. Phytother Res. 30:929–939. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Tiyasatkulkovit W, Malaivijitnond S, Charoenphandhu N, Havill LM, Ford AL and VandeBerg JL: Pueraria mirifica extract and Puerarin enhance proliferation and expression of alkaline phosphatase and type I collagen in primary baboon osteoblasts. Phytomedicine. 21:1498–1503. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Manonai J, Chittacharoen A, Udomsubpayakul U, Theppisai H and Theppisai U: Effects and safety of Pueraria mirifica on lipid profiles and biochemical markers of bone turnover rates in healthy postmenopausal women. Menopause. 15:530–535. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Wang Y, Wang WL, Xie WL, Li LZ, Sun J, Sun WJ and Gong HY: Puerarin stimulates proliferation and differentiation and protects against cell death in human osteoblastic MG-63 cells via ER-dependent MEK/ERK and PI3K/Akt activation. Phytomedicine. 20:787–796. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Wang Y, Ma Y, Zheng Y, Song J, Yang X, Bi C, Zhang D and Zhang Q: In vitro and in vivo anticancer activity of a novel Puerarin nanosuspension against colon cancer, with high efficacy and low toxicity. Int J Pharm. 441:728–735. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Jia L, Hu Y, Yang G and Li P: Puerarin suppresses cell growth and migration in HPV-positive cervical cancer cells by inhibiting the PI3K/mTOR signaling pathway. Exp Ther Med. 18:543–549. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Zhang WG, Liu XF, Meng KW and Hu SY: Puerarin inhibits growth and induces apoptosis in SMMC-7721 hepatocellular carcinoma cells. Mol Med Rep. 10:2752–2758. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Zhang WG, Yin XC, Liu XF, Meng KW, Tang K, Huang FL, Xu G and Gao J: Puerarin induces hepatocellular carcinoma cell apoptosis modulated by MAPK signaling pathways in a dose-dependent manner. Anticancer Res. 37:4425–4431. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Hu Q, Xiang H, Shan J, Jiao Q, Lv S, Li L, Li F, Ren D and Lou H: Two pairs of diastereoisomeric isoflavone glucosides from the roots of Pueraria lobata. Fitoterapia. 144(104594)2020.PubMed/NCBI View Article : Google Scholar | |
|
Shi ZD, Hao L, Han XX, Wu ZX, Pang K, Dong Y, Qin JX, Wang GY, Zhang XM, Xia T, et al: Targeting HNRNPU to overcome cisplatin resistance in bladder cancer. Mol Cancer. 21(37)2022.PubMed/NCBI View Article : Google Scholar | |
|
Nandana PI, Rasyid H, Prihantono P, Yustisia I, Hakim L, Bukhari A and Prasedya ES: Cytotoxicity and apoptosis studies of brucein D against T24 bladder cancer cells. Asian Pac J Cancer Prev. 25:921–930. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Sun Y, Liu X, Tong H, Yin H, Li T, Zhu J, Chen J, Wu L, Zhang X, Gou X and He W: SIRT1 promotes cisplatin resistance in bladder cancer via beclin1 deacetylation-mediated autophagy. Cancers (Basel). 16(125)2023.PubMed/NCBI View Article : Google Scholar | |
|
Mun JY, Baek SW, Jeong MS, Jang IH, Lee SR, You JY, Kim JA, Yang GE, Choi YH, Kim TN, et al: Stepwise molecular mechanisms responsible for chemoresistance in bladder cancer cells. Cell Death Discov. 8(450)2022.PubMed/NCBI View Article : Google Scholar | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Dong J, Guo Y, Ji T, Gao Q, Guan H, Niu Y, Ma Y and Rong S: Effect of Puerariae radix flavones on mRNA expression of N-myc downstream regulatory gene 1 in bladder cancer cell line T24. J Environ Health. 41:941–943. 2024.(In Chinese). | |
|
Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H and Vandenabeele P: Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 15:135–147. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Lavrik IN and Krammer PH: Regulation of CD95/Fas signaling at the DISC. Cell Death Differ. 19:36–41. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Peter ME and Krammer PH: The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ. 10:26–35. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Rehm M, Huber HJ, Dussmann H and Prehn JH: Systems analysis of effector caspase activation and its control by X-linked inhibitor of apoptosis protein. EMBO J. 25:4338–4349. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Cheng J, Zhou T, Liu C, Shapiro JP, Brauer MJ, Kiefer MC, Barr PJ and Mountz JD: Protection from Fas-mediated apoptosis by a soluble form of the Fas molecule. Science. 263:1759–1762. 1994.PubMed/NCBI View Article : Google Scholar | |
|
Kantari C and Walczak H: Caspase-8 and bid: Caught in the act between death receptors and mitochondria. Biochim Biophys Acta. 1813:558–563. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Sartorius U, Schmitz I and Krammer PH: Molecular mechanisms of death-receptor-mediated apoptosis. Chembiochem. 2:20–29. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Kaufmann T, Strasser A and Jost PJ: Fas death receptor signalling: Roles of Bid and XIAP. Cell Death Differ. 19:42–50. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Li H and Lin X: Positive and negative signaling components involved in TNFalpha-induced NF-kappaB activation. Cytokine. 41:1–8. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Wajant H and Scheurich P: TNFR1-induced activation of the classical NF-κB pathway. FEBS J. 278:862–876. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Luo JL, Tan W, Ricono JM, Korchynskyi O, Zhang M, Gonias SL, Cheresh DA and Karin M: Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature. 446:690–694. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Karin M and Lin A: NF-kappaB at the crossroads of life and death. Nat Immunol. 3:221–227. 2002.PubMed/NCBI View Article : Google Scholar | |
|
Ben-Neriah Y and Karin M: Inflammation meets cancer, with NF-κB as the matchmaker. Nat Immunol. 12:715–723. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Baud V and Karin M: Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov. 8:33–40. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Vazquez-Santillan K, Melendez-Zajgla J, Jimenez-Hernandez L, Martínez-Ruiz G and Maldonado V: NF-κB signaling in cancer stem cells: A promising therapeutic target? Cell Oncol (Dordr). 38:327–339. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Cao X and Jin HM: Effects of Fas, NF-κB and caspases on microvascular endothelial cell apoptosis induced by TNFα. Chin J Pathophysiol. 8:15–18. 2001.(In Chinese). |