Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Article in English | MEDLINE | ID: mdl-39091264

ABSTRACT

BACKGROUND: Cancer cachexia-induced skeletal muscle fibrosis (SMF) impairs muscle regeneration, alters the muscle structure and function, reduces the efficacy of anticancer drugs, diminishes the patient's quality of life and shortens overall survival. RUNX family transcription factor 2 (Runx2), a transcription factor, and collagen type I alpha 1 chain (COL1A1), the principal constituent of SMF, have been linked previously, with Runx2 shown to directly modulate COL1A1 mRNA levels. l-Carnitine, a marker of cancer cachexia, can alleviate fibrosis in liver and kidney models; however, its role in cancer cachexia-associated fibrosis and the involvement of Runx2 in the process remain unexplored. METHODS: Female C57 mice (48 weeks old) were inoculated subcutaneously with MC38 cells to establish a cancer cachexia model. A 5 mg/kg dose of l-carnitine or an equivalent volume of water was administered for 14 days via oral gavage, followed by assessments of muscle function (grip strength) and fibrosis. To elucidate the interplay between the deltex E3 ubiquitin ligase 3L(DTX3L)/Runx2/COL1A1 axis and fibrosis in transforming growth factor beta 1-stimulated NIH/3T3 cells, a suite of molecular techniques, including quantitative real-time PCR, western blot analysis, co-immunoprecipitation, molecular docking, immunofluorescence and Duolink assays, were used. The relevance of the DTX3L/Runx2/COL1A1 axis in the gastrocnemius was also explored in the in vivo model. RESULTS: l-Carnitine supplementation reduced cancer cachexia-induced declines in grip strength (>88.2%, P < 0.05) and the collagen fibre area within the gastrocnemius (>57.9%, P < 0.05). At the 5 mg/kg dose, l-carnitine also suppressed COL1A1 and alpha-smooth muscle actin (α-SMA) protein expression, which are markers of SMF and myofibroblasts. Analyses of the TRRUST database indicated that Runx2 regulates both COL1A1 and COL1A2. In vitro, l-carnitine diminished Runx2 protein levels and promoted its ubiquitination. Overexpression of Runx2 abolished the effects of l-carnitine on COL1A1 and α-SMA. Co-immunoprecipitation, molecular docking, immunofluorescence and Duolink assays confirmed an interaction between DTX3L and Runx2, with l-carnitine enhancing this interaction to promote Runx2 ubiquitination. l-Carnitine supplementation restored DTX3L levels to those observed under non-cachectic conditions, both in vitro and in vivo. Knockdown of DTX3L abolished the effects of l-carnitine on Runx2, COL1A1 and α-SMA in vitro. The expression of DTX3L was negatively correlated with the levels of Runx2 and COL1A1 in untreated NIH/3T3 cells. CONCLUSIONS: This study revealed a previously unrecognized link between Runx2 and DTX3L in SMF and demonstrated that l-carnitine exerted a significant therapeutic impact on cancer cachexia-associated SMF, potentially through the upregulation of DTX3L.

2.
Sci Total Environ ; 935: 173430, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38782273

ABSTRACT

The prevalence of pollen allergies is a pressing global issue, with projections suggesting that half of the world's population will be affected by 2050 according to the estimation of the World Health Organization (WHO). Accurately forecasting pollen allergy risks requires identifying key factors and their thresholds for aerosol pollen. To address this, we developed a technical framework combining advanced machine learning and SHapley Additive exPlanations (SHAP) technology, focusing on Beijing. By analyzing meteorological data and vegetation phenology, we identified the factors influencing next-day's pollen concentration (NDP) in Beijing and their thresholds. Our results highlight vegetation phenology data from Synthetic Aperture Radar (SAR), temperature, wind speed, and atmospheric pressure as crucial factors in spring. In contrast, the Normalized Difference Vegetation Index (NDVI), air temperature, and wind speed are significant in autumn. Leveraging SHAP technology, we established season-specific thresholds for these factors. Our study not only confirms previous research but also unveils seasonal variations in the relationship between radar-derived vegetation phenology data and NDP. Additionally, we observe seasonal fluctuations in the influence patterns and threshold values of daily air temperatures on NDP. These insights are pivotal for improving pollen concentration prediction accuracy and managing allergic risks effectively.


Subject(s)
Air Pollutants , Allergens , Environmental Monitoring , Machine Learning , Pollen , Seasons , Air Pollutants/analysis , Environmental Monitoring/methods , Allergens/analysis , Beijing , Air Pollution/statistics & numerical data
4.
Clin Nutr ; 42(6): 1048-1058, 2023 06.
Article in English | MEDLINE | ID: mdl-37178592

ABSTRACT

BACKGROUND & AIMS: The present study aimed to compare the ability of the GLIM criteria, PG-SGA and mPG-SGA to diagnose malnutrition and predict survival among Chinese lung cancer (LC) patients. METHODS: This was a secondary analysis of a multicenter, prospective, nationwide cohort study, 6697 LC inpatients were enrolled between July 2013 and June 2020. The sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), area under the curve (AUC), and quadratic weighted Kappa coefficients were calculated to compare the ability to diagnose malnutrition. There were 754 patients who underwent follow-up for a median duration of 4.5 years. The associations between the nutritional status and survival were analyzed by the Kaplan-Meier method and multivariable Cox proportional hazard regression models. RESULTS: The median age of LC patients was 60 (53, 66), and 4456 (66.5%) were male. There were 617 (9.2%), 752 (11.2%), 1866 (27.9%), and 3462 (51.7%) patients with clinical stage Ⅰ, Ⅱ, Ⅲ, and Ⅳ LC, respectively. Malnutrition was present in 36.1%-54.2% (as evaluated using different tools). Compared with the PG-SGA (used as the diagnostic reference), the sensitivity of the mPG-SGA and GLIM was 93.7% and 48.3%; the specificity was 99.8% and 78.4%; and the AUC was 0.989 and 0.633 (P < 0.001). The weighted Kappa coefficients were 0.41 for the PG-SGA vs. GLIM, 0.44 for the mPG-SGA vs. GLIM, and 0.94 for the mPG-SGA vs PG-SGA in patients with stage Ⅰ-Ⅱ LC. These values were respectively 0.38, 0.39, and 0.93 in patients with stage Ⅲ-Ⅳ of LC. In a multivariable Cox analysis, the mPG-SGA (HR = 1.661, 95%CI = 1.348-2.046, P < 0.001), PG-SGA (HR = 1.701, 95%CI = 1.379-2.097, P < 0.001) and GLIM (HR = 1.657, 95%CI = 1.347-2.038, P < 0.001) showed similar death hazard ratios. CONCLUSIONS: The mPG-SGA provides nearly equivalent power to predict the survival of LC patients as the PG-SGA and the GLIM, indicating that all three tools are applicable for LC patients. The mPG-SGA has the potential to be an alternative replacement for quick nutritional assessment among LC patients.


Subject(s)
Lung Neoplasms , Malnutrition , Humans , Male , Female , Cohort Studies , Prospective Studies , Malnutrition/diagnosis , Inpatients , Lung Neoplasms/complications , Lung Neoplasms/diagnosis , Nutritional Status , Nutrition Assessment
5.
J Cachexia Sarcopenia Muscle ; 14(1): 517-533, 2023 02.
Article in English | MEDLINE | ID: mdl-36567070

ABSTRACT

BACKGROUND: The fat mass and nutritional status play important roles in the onset and progression of cancer cachexia. The present study evaluated the joint prognostic value of the fat mass, as indicated by the triceps skinfold thickness (TSF), and the serum albumin level, for mortality in patients with cancer cachexia. METHODS: We performed a multicentre cohort study including 5134 patients with cancer cachexia from January 2013 to April 2019. The sum of the TSF (mm) and serum albumin (g/L) was defined as the triceps skinfold-albumin index (TA). Harrell's C index, a time-dependent receiver operating characteristic (ROC) curve analysis and the area under the curve (AUC) were used to evaluate the prognostic performance of the TA and other indices. Optimal stratification was used to identify the thresholds to define a low TA, and the association of the TA with all-cause mortality was evaluated using Kaplan-Meier analysis and Cox proportional hazard regression models. RESULTS: The study enrolled 2408 women and 2726 men with a median age of 58.6 years and a median follow-up of 44 months. A total of 607 women (TA < 49.9) and 817 men (TA < 45.6) were classified as having a low TA. The TA showed better discrimination performance (C index = 0.621, 95% confidence interval [CI] = 0.607-0.636) to predict mortality in patients with cancer cachexia than the handgrip strength, the nutritional risk index, the prognostic nutritional index, the controlling nutritional status index, the systemic immune-inflammation index, the modified Glasgow prognostic score, and the TSF or albumin alone in the study population (all P < 0.05). The 1-, 3- and 5-year time-dependent ROC analyses (AUC = 0.647, 0.625 and 0.630, respectively) showed that the TA had the highest prognostic value among all indices investigated (all P < 0.05). Univariate analysis showed that a lower TA was associated with an increased death hazard (hazard ratio [HR] = 1.859, 95% CI = 1.677-2.062), regardless of the sex and cancer type. Multivariable survival analysis showed that a lower TA was independently associated with an increased death hazard (HR = 1.381, 95% CI = 1.223-1.560). This association was significantly strengthened in patients who did not receive curative chemotherapy (HR = 1.491, 95% CI = 1.298-1.713), those who had higher serum total protein levels (HR = 1.469, 95% CI = 1.284-1.681) and those with better physical performance (HR = 1.453, 95% CI = 1.271-1.662). CONCLUSIONS: This study defined and evaluated a new prognostic index, the TA, which may improve the selection of intervention strategies to optimize the survival of patients with cancer cachexia.


Subject(s)
Cachexia , Neoplasms , Male , Humans , Female , Middle Aged , Cohort Studies , Cachexia/diagnosis , Cachexia/etiology , Hand Strength , Prognosis , Neoplasms/complications , Serum Albumin/analysis , Serum Albumin/metabolism
6.
Support Care Cancer ; 31(1): 72, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36543973

ABSTRACT

BACKGROUND: Early recognition of cachexia is essential for ensuring the prompt intervention and treatment of cancer patients. However, the diagnosis of cancer cachexia (CC) usually is delayed. This study aimed to establish an accurate and high-efficiency diagnostic system for CC. METHODS: A total of 4834 cancer inpatients were enrolled in the INSCOC project from July 2013 to June 2020. All cancer patients in the study were randomly assigned to a development cohort (n=3384, 70%) and a validation cohort (n=1450, 30%). The least absolute shrinkage and selection operator (LASSO) method and multivariable logistic regression were used to identify the independent predictors for developing the dynamic nomogram. Discrimination and calibration were adopted to evaluate the ability of nomogram. A decision curve analysis (DCA) was used to evaluate clinical use. RESULTS: We combined 5 independent predictive factors (age, NRS2002, PG-SGA, QOL by the QLQ-C30, and cancer categories) to establish the online dynamic nomogram system. The C-index, sensitivity, and specificity of the nomo-system to predict CC was 0.925 (95%CI, 0.916-0.934, P < 0.001), 0.826, and 0.862 in the development set, while the values were 0.923 (95%CI, 0.909-0.937, P < 0.001), 0.854, and 0.829 in the validation set. In addition, the calibration curves of the diagnostic nomogram also presented good agreement with the actual situation. DCA showed that the model is clinically useful and can increase the clinical benefit in cancer patients. CONCLUSIONS: This study developed an online dynamic nomogram system with outstanding accuracy to help clinicians and dieticians estimate the probability of cachexia. This simple-to-use online nomogram can increase the clinical benefit in cancer patients and is expected to be widely adopted.


Subject(s)
Cachexia , Neoplasms , Humans , Cachexia/diagnosis , Cachexia/etiology , Cohort Studies , Inpatients , Nomograms , Quality of Life , China , Neoplasms/complications
7.
Clin Nutr ; 41(6): 1208-1217, 2022 06.
Article in English | MEDLINE | ID: mdl-35504163

ABSTRACT

BACKGROUND & AIMS: Although malnutrition remains a global public health concern, and has proved to be a major contributor to death and illness, there has been a foundational lack of a gold standard for diagnostic testing for clinical application. The Global Leadership Initiative on Malnutrition (GLIM) criteria were established to normalize the diagnosis of malnutrition, but their use remains controversial. Therefore, we carried out a meta-analysis based on the published literature to assess the accuracy of the GLIM criteria for diagnosing malnutrition. METHODS: We utilized publication databases (including CENTRAL, MEDLINE, and EMBASE) to acquire research studies published from the initial use of the GLIM criteria in 2019 until January 22, 2022 that used the criteria to diagnose malnutrition. We conducted this meta-analysis with reference to the recommendations from the PRISMA-DTA statement. We separately calculated the amalgamated sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and AUC with 95%CI for the GLIM criteria. Then, we aggregated and presented the data by drawing forest plots to assess the real accuracy of the criteria. A subgroup analysis was also carried out to identify the potential sources of heterogeneity. RESULTS: After the initial search of the CENTRAL, EMBASE, and MEDLINE databases, a total of 451 unique studies were identified. Twenty studies met our selection standards and 10,781 total patients were included in the meta-analysis. We noted that 4761 of the 10,781 patients (44.2%) were malnourished. The amalgamated sensitivity of the GLIM criteria was 0.72 (95%CI, 0.64-0.78), the specificity was 0.82 (95%CI, 0.72-0.88), the PLR was 3.9 (95%CI, 2.6-6.1), NLR was 0.35 (95%CI, 0.27-0.44), DOR was 11 (95%CI, 6-20), and AUC was 0.82 (95%CI, 0.79-0.85). Based on the results of a subgroup analysis using the SGA as a reference standard, the GLIM criteria had better diagnostic value (sensitivity, 0.81; specificity, 0.80; DOR, 17; AUC, 0.87). CONCLUSIONS: The GLIM criteria have high diagnostic accuracy for distinguishing patients with malnutrition, and the GLIM criteria seem to have the potential to be used as a gold standard for diagnosing malnutrition in clinical practice. Moreover, the subgroup analysis showed a better diagnostic value for the GLIM criteria compared to the SGA used as a reference standard. Large-scale diagnostic trials and additional refinements to simplify the criteria are urgently needed to increase the clinical utilization of the GLIM criteria in the future.


Subject(s)
Malnutrition , Nutrition Assessment , Humans , Leadership , Malnutrition/diagnosis , Nutritional Status , Odds Ratio
8.
J Food Biochem ; 46(5): e14063, 2022 05.
Article in English | MEDLINE | ID: mdl-35128673

ABSTRACT

Mounting evidence has linked both obesity and metabolic disorders with dysbiosis of the gut microbiota. Dietary inulin is conducive to modulating this dysbiosis, and represents a potential means to improve disorders of glucose and lipid metabolism. However, the mechanisms underlying these improvements are largely unclear. Obese ob/ob mice were fed a standard chow, a low fiber diet (LFD) or a high fiber diet (HFD) for 4 weeks, and the body weight, fecal short chain fatty acids (SCFAs) level, and plasma and liver lipid profiles were analyzed. Oral glucose tolerance testing, and gut microbiota sequencing were also conducted. Dietary inulin improved the dysbiosis of the gut microbiota, attenuated the decrease in phylum Bacteroidetes, repressed the increase of phylum Firmicutes, and led to an increase in the ratio of Firmicutes/Bacteroidetes. At the family level, inulin promoted the expansion of SCFAs-producing Ruminococcaceae and Lachnospiraceae bacteria, which increased the fecal SCFAs concentrations. At the genus level, inulin increased the levels of Bacteroides and Bifidobacteria. Furthermore, our results revealed that there was enhanced expression of angiopoietin-like protein 4 (ANGPTL4), which might be induced by the higher production of SCFAs, and this may underlie the improvements in the disorders of glucose and lipid metabolism seen in mice with added dietary inulin. In conclusion, inulin may ameliorate metabolic disorders by remodeling the gut microbiota and increasing the production of SCFAs, which might be mediated by the ANGPTL4-related signaling pathway. Interventions targeting the gut microbiota warrant further investigation as a novel therapy for metabolic diseases. PRACTICAL APPLICATIONS: Mounting evidence has linked both obesity and metabolic disorders with dysbiosis of the gut microbiota. Dietary inulin is conducive to modulating this dysbiosis, and represents a potential means to improve disorders of glucose and lipid metabolism. However, the mechanisms underlying these improvements are largely unclear. In the present study, we investigated the effects of dietary fiber (inulin) on metabolic homeostasis using ob/ob mice. The results of our study demonstrate that inulin-induced remodeling of the gut microbiota resulted in increased production of short chain fatty acids (SCFAs), leading to the enhanced expression of angiopoietin-like protein 4 (ANGPTL4), which improved the glucose and lipid metabolism. Our results suggest that the gut microbiota, SCFAs and ANGPTL4 pathway at least partially mediate the beneficial effects of inulin on metabolic disorders in ob/ob mice.


Subject(s)
Gastrointestinal Microbiome , Metabolic Diseases , Angiopoietin-Like Protein 4 , Animals , Diet, High-Fat , Dysbiosis/drug therapy , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Firmicutes , Glucose , Inulin/pharmacology , Mice , Obesity/metabolism
9.
Nutr Metab (Lond) ; 18(1): 98, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34724970

ABSTRACT

BACKGROUND: Recent studies suggest potential benefits of applying L-carnitine in the treatment of cancer cachexia, but the precise mechanisms underlying these benefits remain unknown. This study was conducted to determine the mechanism by which L-carnitine reduces cancer cachexia. METHODS: C2C12 cells were differentiated into myotubes by growing them in DMEM for 24 h (hrs) and then changing the media to DMEM supplemented with 2% horse serum. Differentiated myotubes were treated for 2 h with TNF-α to establish a muscle atrophy cell model. After treated with L-carnitine, protein expression of MuRF1, MaFbx, FOXO3, p-FOXO3a, Akt, p-Akt, p70S6K and p-p70S6K was determined by Western blotting. Then siRNA-Akt was used to determine that L-carnitine ameliorated cancer cachexia via the Akt/FOXO3/MaFbx. In vivo, the cancer cachexia model was established by subcutaneously transplanting CT26 cells into the left flanks of the BALB/c nude mice. After treated with L-carnitine, serum levels of IL-1, IL-6 and TNF-α, and the skeletal muscle content of MuRF1, MaFbx, FOXO3, p-FOXO3a, Akt, p-Akt, p70S6K and p-p70S6K were measured. RESULTS: L-carnitine increased the gastrocnemius muscle (GM) weight in the CT26-bearing cachexia mouse model and the cross-sectional fiber area of the GM and myotube diameters of C2C12 cells treated with TNF-α. Additionally, L-carnitine reduced the protein expression of MuRF1, MaFbx and FOXO3a, and increased the p-FOXO3a level in vivo and in vitro. Inhibition of Akt, upstream of FOXO3a, reversed the effects of L-carnitine on the FOXO3a/MaFbx pathway and myotube diameters, without affecting FOXO3a/MuRF-1. In addition to regulating the ubiquitination of muscle proteins, L-carnitine also increased the levels of p-p70S6K and p70S6K, which are involved in protein synthesis. Akt inhibition did not reverse the effects of L-carnitine on p70S6K and p-p70S6K. Hence, L-carnitine ameliorated cancer cachexia via the Akt/FOXO3/MaFbx and p70S6K pathways. Moreover, L-carnitine reduced the serum levels of IL-1 and IL-6, factors known to induce cancer cachexia. However, there were minimal effects on TNF-α, another inducer of cachexia, in the in vivo model. CONCLUSION: These results revealed a novel mechanism by which L-carnitine protects muscle cells and reduces inflammation related to cancer cachexia.

10.
Front Oncol ; 11: 648985, 2021.
Article in English | MEDLINE | ID: mdl-34026624

ABSTRACT

Castration-resistant (androgen-independent) and PTEN-deficient prostate cancer is a challenge in clinical practice. Sorafenib has been recommended for the treatment of this type of cancer, but is associated with several adverse effects. Platycodin D (PD) is a triterpene saponin with demonstrated anti-cancer effects and a good safety profile. Previous studies have indicated that PC3 cells (PTEN -/-, AR -/-) are sensitive to PD, suggesting that it may also be a useful treatment for castration-resistance prostate cancer. We herein investigated the effects of combining PD with sorafenib to treat PTEN-deficient prostate cancer cells. Our data show that PD promotes sorafenib-induced apoptosis and cell cycle arrest in PC3 cells. Of interest, PD only promoted the anti-cancer effects of sorafenib in Akt-positive and PTEN-negative prostate cancer cells. Mechanistic studies revealed that PD promoted p-Akt ubiquitination by increasing the p-Akt level. PD also increased the protein and mRNA expression of FOXO3a, the downstream target of Akt. Meanwhile, PD promoted the activity of FOXO3a and increased the protein expression of Fasl, Bim and TRAIL. Interestingly, when FOXO3a expression was inhibited, the antitumor effects of both PD and sorafenib were individually inhibited, and the more potent effects of the combination treatment were inhibited. Thus, the combination of PD and sorafenib may exert potent anti-cancer effects specifically via FOXO3a. The use of Akt inhibitors or FOXO3a agonists, such as PD, may represent a promising approach for the treatment of androgen-independent and PTEN-deficient prostate cancer.

11.
Int J Oncol ; 56(2): 439-447, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894265

ABSTRACT

Ophiopogonin D' (OPD') is a natural compound extracted from Ophiopogon japonicus, which is a plant used in traditional Chinese medicine. Our previous study has indicated that OPD' exhibits antitumor activity against androgen­independent prostate cancer (PCa), but the effects and the underlying molecular mechanism of action of OPD' in androgen­dependent PCa were unclear. In the present study, OPD' induced significant necroptosis in androgen­dependent LNCaP cancer cells by activating receptor­interacting serine/threonine­protein kinase 1 (RIPK1). Exposure to OPD' also increased Fas ligand (FasL)­dependent RIPK1 protein expression. The OPD'­induced necroptosis was inhibited by a RIPK1 inhibitor necrostatin­1, further supporting a role for RIPK1 in the effects of OPD´. The antitumor effects of OPD' were also inhibited by a mixed lineage kinase domain­like protein (MLKL) inhibitor necrosulfonamide. Following treatment with inhibitors of RIPK1 and MLKL, the effects of OPD' on LNCaP cells were inhibited in an additive manner. In addition, co­immunoprecipitation assays demonstrated that OPD' induced RIPK3 upregulation, leading to the assembly of a RIPK3­MLKL complex, which was independent of RIPK1. Furthermore, OPD' increased the expression of Fas­associated death domain, which is required to induce necroptosis in LNCaP cells. OPD' also regulated the expression levels of FasL, androgen receptor and prostate­specific antigen in a RIPK1­dependent manner. These results suggested that OPD' may exhibit potential as an anti­PCa agent by inducing RIPK1­ and MLKL­dependent necroptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Necroptosis/drug effects , Prostatic Neoplasms/drug therapy , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Saponins/pharmacology , Spirostans/pharmacology , Acrylamides/pharmacology , Androgens/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Male , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , RNA, Small Interfering , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Saponins/therapeutic use , Spirostans/therapeutic use , Sulfonamides/pharmacology , Up-Regulation/drug effects
12.
Prostate ; 79(1): 62-72, 2019 01.
Article in English | MEDLINE | ID: mdl-30242862

ABSTRACT

BACKGROUND: Voltage-gated sodium channels (VGSCs) are involved in several cellular processes related to cancer cell growth and metastasis, including adhesion, proliferation, apoptosis, migration, and invasion. We here in investigated the effects of S0154 and S0161, two novel synthetic sodium channel blockers (SCBs), on human prostate cancer cells (PC3, DU145, and LnCaP) and a prostate cancer xenograft model. METHODS: The MTT assay was used to assess the anticancer effects of SCBs in PC3, DU145, and LnCaP cells. Sodium indicator and glucose uptake assays were used to determine the effects of S0154 and S0161 in PC3 cells. The impact of these SCBs on the proliferation, cell cycle, apoptosis, migration, and invasion of PC3 cells were determined using a CFDA-SE cell proliferation assay, cell cycle assay, annexin V-FITC apoptosis assay, transwell cell invasion assay, and wound-healing assay, respectively. The protein expression levels of Nav1.6, Nav1.7, CDK1, cyclin B1, MMP2, MMP9 in PC3 cells were analysis by Western blotting. The in vivo anticancer activity was evaluated using a PC3 xenograft model in nude mice. RESULTS: S0154 and S0161 both showed anticancer and anti-metastatic effects against prostate cancer cells and significantly inhibited cell viability, with IC50 values in the range of 10.51-26.60 µmol/L (S0154) and 5.07-11.92 µmol/L (S0161). Both compounds also increased the intracellular level of sodium, inhibited the protein expression of two α subunits of VGSCs (Nav1.6 and Nav1.7), and caused G2/M phase cell cycle arrest, with no or minor effects on cell apoptosis. Concentrations of 5 and 10 µmol/L of S0154 and S0161 significantly decreased the glucose uptake of PC3 cells. The compounds also inhibited the proliferation of PC3 cells and decreased their invasion in transwell assays. Furthermore, S0161 exerted antitumor activity in an in vivo PC3 xenograft model in nude mice, inhibiting the growth of the tumors by about 51% compared to the control group. CONCLUSIONS: These results suggest that S0154 and S0161 have anticancer and anti-metastasis effects in prostate cancer cells both in vitro and in vivo, supporting their further development as potential therapeutic agents for prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Prostatic Neoplasms/drug therapy , Sodium Channel Blockers/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/physiology , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays/methods
13.
Front Pharmacol ; 9: 432, 2018.
Article in English | MEDLINE | ID: mdl-29760660

ABSTRACT

Objective: The purpose of this study was to evaluate the anticancer effects of Ophiopogonin D' (OPD', a natural product extracted from a traditional Chinese medicine (Radix Ophiopogonis) against androgen-independent prostate cancer cells and to explore the underlying molecular mechanism(s) of action. Methods: The CCK-8 assay was used to assess the viability of prostate cancer cells. The cell morphology was examined by an ultrastructural analysis via transmission electron microscopy. Cells in apoptosis (early and late stages) were detected using an Annexin V-FITC/propidium iodide kit with a FACSCaliber flow cytometer. JC-1, a cationic lipophilic probe, was employed to measure the mitochondrial membrane potential (MMP) of PC3 cells. Changes in the protein expression of RIPK1, C-RIPK1, caspase 8, cleaved-caspase 8, Bim, Bid, caspase 10, and cleaved-caspase 10 were evaluated by Western blotting. The mRNA expression of Bim was examined by quantitative real-time reverse transcription polymerase chain reaction. Z-VAD-FMK (a caspase inhibitor) and necrostatin-1 (a specific inhibitor of RIPK1) were utilized to determine whether the cell death was mediated by RIPK1 or caspases. PC3 and DU145 xenograft models in BALB/c nude mice were used to evaluate the anticancer activity of OPD' in vivo. Results: OPD' was shown to exert potent anti-tumor activity against PC3 cells. It induced apoptosis via a RIPK1-related pathway, increased the protein expression levels of RIPK1 and Bim, and decreased the levels of cleaved-RIPK1, caspase 8, cleaved-caspase 8, Bid, caspase 10, and cleaved-caspase 10. OPD' also increased the mRNA expression of Bim. The protein expression of Bim was decreased when cells were pre-treated with necrostatin-1. Treatment with OPD' inhibited the growth of PC3 and DU145 xenograft tumors in BALB/c nude mice. Conclusion: OPD' significantly inhibited the in vitro and in vivo growth of prostate cells via RIPK1, suggesting that OPD' may be developed as a potential anti-prostate cancer agent.

14.
Oncol Rep ; 36(3): 1447-56, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27432230

ABSTRACT

The objective of the present study was to explore the in vitro and in vivo anticancer effects of Platycodin D (PD), derived from Platycodin grandiflorum, on highly metastatic MDA-MB-231 breast cancer cells. Using the MTT assay, we found that PD inhibited MDA-MB-231 cell growth in a concentration-dependent manner, with an IC50 value of 7.77±1.86 µM. Further studies showed that PD had anti-proliferative effects and induced cell cycle arrest in the G0/G1 phase. To explore the detailed mechanism(s) by which PD suppressed MDA-MB-231 cell growth, western blot analyses were used to detect the expression levels of proteins related to cell proliferation and survival. The data showed that PD decreased the expression of proteins related to the G0/G1 phases, downregulated the protein expression of MDM2, MDMX, and mutant p53, and increased the expression levels of p21 and p27 in vitro. We verified the effects of PD on the expression of MDM2, MDMX, mutant p53, p21 and p27 using a pcDNA3-Flag-MDM2 plasmid and MDM2 siRNA transfection, and found that PD inhibited MDA-MB-231 cell viability by targeting MDM2 and mutant p53. Compared with the corresponding parental cells, the cells with siRNA-MDM2 transfection had a greater decrease in cell viability and proliferation, while those with pcDNA3-MDM2 plasmid transfection did not show any increase in the effects of PD. We also established a MDA-MB-231 xenograft model in BALB/c nude mice, and found that PD significantly inhibited the growth of MDA-MB-231 xenograft tumors in these mice. The expression levels of various proteins in the tumor tissue exhibited changes similar to those observed in vitro. These findings indicate that PD exerted in vitro and in vivo anticancer effects against MDA-MB-231 breast cancer cells, that PD is a potential MDM2/MDMX inhibitor, and that the anticancer effects of PD were likely associated with its inhibition of these proteins. Our observations help to identify a mechanism by which PD functions as an anti-breast cancer agent.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Oncogenes/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , Saponins/metabolism , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Female , G1 Phase/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Resting Phase, Cell Cycle/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
15.
Curr Cancer Drug Targets ; 16(5): 455-65, 2016.
Article in English | MEDLINE | ID: mdl-26638886

ABSTRACT

Forkhead box O3 (FOXO3a) is a transcription factor with tumor suppressor functions that plays an important role in prostate cancer. Daidzein, one of the soy isoflavones present in soy-based foods, has been shown to exert anti-tumor effects in vitro and in vivo. We herein investigated the inhibitory effects of S-equol, an isoflavandiol metabolized from daidzein by bacterial flora in the intestines, on the LnCaP, DU145 and PC3 human prostate cancer cell lines. Our results showed that S-equol and R-equol inhibited the growth of all three cell lines. Additional studies revealed that S-equol caused cell cycle arrest in the G2/M phase in PC3 cells by downregulating Cyclin B1 and CDK1 and upregulating CDK inhibitors (p21 and p27), as well as inducing apoptosis by upregulating Fas ligand (FasL) and the expression of proapoptotic Bim. Additionally, S-equol increased the expression of FOXO3a, decreased the expression of p-FOXO3a and enhanced the nuclear stability of FOXO3a. S-equol also decreased the expression of MDM2, which serves as an E3 ubiquitin ligase for p-FOXO3a, thus preventing p-FOXO3a degradation by the proteasome. Mechanistic studies showed that S-equol targeted the Akt/FOXO3a pathway, which is important for prostate cancer cell survival, cell cycle progression and apoptosis. Moreover, treatment with S-equol inhibited the growth of PC3 xenograft tumors in BALB/c nude mice. Overall, the data from the present study demonstrate that S-equol has significant anti-prostate cancer activities in vitro and in vivo, and indicate that its anticancer effects were likely associated with the activation of FOXO3a via an Akt-specific pathway and inhibitory effects on MDM2 expression. The results not only provide a better understanding of the molecular mechanisms of this unique secondary metabolite of a natural anti-cancer compound, but also provide a basis for the development of daidzein and its analogs as novel anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Equol/pharmacology , Forkhead Box Protein O3/metabolism , Isoflavones/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Biological Products/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects
16.
Curr Cancer Drug Targets ; 14(9): 860-71, 2015.
Article in English | MEDLINE | ID: mdl-25431082

ABSTRACT

Platycodin D (PD), a major saponin derived from Platycodin grandiflorum, exerted cytotoxicity against prostate cancer cell lines (PC3, DU145 and LNCaP cells) with IC50 values in the range of 11.17 to 26.13 µmol/L, whereas RWPE-1 cells (a non-malignant human prostate epithelial cell line) were not significantly affected. A further study in these cell lines showed that PD could potently affect cell proliferation (indicated by the bromodeoxyuridine assay), induce cell apoptosis (determined by Annexin V-FITC flow cytometry) and cause cell cycle arrest (indicated by PI staining). After being treated with PD for 48 hours, DU145 and LNCaP cells were arrested in the G0 /G1 phase, and PC3 cells were arrested in the G2/M phase. A Western blotting analysis indicated that PD increased the expression of the FOXO3a transcription factor, decreased the expression of p-FOXO3a and MDM2 and increased the expression of FOXO-responsive genes, p21 and p27. MDM2 silencing (transiently by siRNA-MDM2) increased the PD-induced FOXO3a protein expression, while MDM2 overexpression (in cells transiently transfected with a pcDNA3-MDM2 plasmid) decreased the PD-induced expression of the FOXO3a protein. Moreover, PD dose-dependently inhibited the growth of PC3 xenograft tumors in BALB/c nude mice. A Western blotting analysis of the excised xenograft tumors indicated that similar changes in protein expression also occurred in vivo. These results suggest that PD exhibits significant activity against prostate cancer in vitro and in vivo. The FOXO3a transcription factor appears to be involved in the activity of PD. Together, all of these findings provide a basis for the future development of this agent for human prostate cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Forkhead Transcription Factors/agonists , Gene Expression Regulation, Neoplastic/drug effects , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Saponins/therapeutic use , Triterpenes/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line , Dose-Response Relationship, Drug , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Silencing , Humans , Male , Mice, Nude , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Resting Phase, Cell Cycle/drug effects , Saponins/administration & dosage , Saponins/adverse effects , Saponins/pharmacology , Specific Pathogen-Free Organisms , Triterpenes/administration & dosage , Triterpenes/adverse effects , Triterpenes/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
PLoS One ; 8(11): e82068, 2013.
Article in English | MEDLINE | ID: mdl-24303079

ABSTRACT

BACKGROUND: The extract of Platycodon grandiflorum has been reported to have effective spermicidal activity. This study was designed to evaluate the spermicidal and contraceptive activity, as well as the safety, of Platycodin D (PD), a major saponin in Platycodon grandiflorum. METHODS: Using the computer-aided sperm analysis (CASA) test criteria, the sperm-immobilizing activity of PD was studied using highly motile human sperm. The sperm viability was assessed by fluorescent staining using SYBR-14 (living sperm) and propidium iodide (dead sperm). The sperm membrane integrity was assessed by evaluating the hypo-osmotic swelling (HOS) and examinations by transmission electron microscopy (TEM) and scanning electron microscopy (SEM). The in vivo contraceptive efficacy was evaluated in rats using post-intrauterine PD application. The comet assay was employed to determine whether PD caused DNA damage in the sperm. Vaginal biopsies were also performed to determine whether the PD gel induced vaginal inflammation. RESULTS: A dose-dependent effect of PD on the sperm motility and viability was observed. The maximum spermicidal effect was observed with a 0.25 mM concentration of PD. More than 70% of the PD-treated sperm lost their HOS responsiveness at a concentration of 0.20 mM PD, indicating that PD caused injury to the sperm plasma membrane. TEM and SEM revealed significant damage to both the head and tail membranes of the sperm. PD decreased the fertility to zero in rats, was non-DNA damaging and was not harmful to the vaginal tissue in the rats. CONCLUSION: PD has significant spermicidal activity that should be explored in further studies.


Subject(s)
Platycodon/chemistry , Saponins/pharmacology , Spermatocidal Agents/pharmacology , Triterpenes/pharmacology , Adult , Animals , Cell Survival/drug effects , Comet Assay , Contraception , Female , Humans , Male , Mucous Membrane/drug effects , Rats , Saponins/toxicity , Sperm Motility/drug effects , Spermatocidal Agents/toxicity , Spermatozoa/cytology , Spermatozoa/ultrastructure , Triterpenes/toxicity , Vagina/drug effects , Young Adult
19.
Clin Exp Hypertens ; 32(8): 491-8, 2010.
Article in English | MEDLINE | ID: mdl-21091365

ABSTRACT

Several previous trials from Western population studies have showed that statins may help reduce blood pressure (BP). However, randomized clinical data is limited. Xuezhikang, a partially extract of red yeast rice, contains a family of naturally occurring statins, and has a marked impact on lipids, but it is unknown whether Xuezhikang has any effect on BP during long-term follow-up in the Chinese population. This is a post-hoc subgroup analysis of a randomized, double-blinded, placebo-controlled, parallel group clinical trial, Chinese Coronary Secondary Prevention Study (CCSPS). A total of 2704 hypertensive patients with previous myocardial infarction (MI) were assigned either to placebo (n = 1341) or to Xuezhikang (n = 1363) daily for an average of 4.5 years. The primary outcome was the unadjusted changes in mean arterial pressure (MAP) from baseline to 6 months. We also assessed systolic blood pressure (SBP), diastolic blood pressure (DBP), and pulse pressure. Analysis of covariance was used to calculate the adjusted effects of treatment on changes in these outcomes at 6, 12, 24, and 48 months post-randomization, after controlling for potential confounders. This analysis included 2704/4870 (55.5%) hypertensive patients for whom BP was measured at baseline and at least one follow-up visit after randomization. Median duration of the follow-up was 4.5 years (54 months), and 25 patients (0.92%) were lost to the last follow-up because of adverse effects. The results showed that the unadjusted and adjusted changes in MAP, SBP, DBP, or pulse pressure from baseline were not significantly different for Xuezhikang or placebo recipients at 6, 12, 24, and 48 months after randomization. In this post-hoc subgroup analysis, we failed to demonstrate any significant reducing effects of Xuezhikang on BP in Chinese hypertensive patients with previous MI, suggesting that further prospective study on the effects of statins on BP would be needed, especially in high-risk patients.


Subject(s)
Antihypertensive Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Hypertension/drug therapy , Myocardial Infarction/complications , Adolescent , Adult , Aged , Blood Pressure/drug effects , China , Female , Follow-Up Studies , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertension/complications , Hypertension/physiopathology , Lipids/blood , Male , Middle Aged , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Young Adult
20.
Ann Med ; 42(3): 231-40, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20350253

ABSTRACT

BACKGROUND: The lowering of cholesterol concentrations in individuals at high risk for cardiovascular disease improves clinical outcome. Xuezhikang has a marked impact on lipids. METHODS: In this randomized, double-blinded, placebo-controlled, parallel-group clinical trial, a total of 2704 hypertensive patients with previous myocardial infarction (MI) were assigned either to placebo (n = 1341) or to Xuezhikang (0.6 g twice daily, n = 1363) for an average of 4.5 years. The primary end-point was recurrent coronary events; the secondary end-point was all-cause mortality and other clinical events, including adverse effects. RESULTS: There were no differences between the Xuezhikang and placebo group in base-line characteristics. However, Xuezhikang treatment reduced the incidence of coronary events by 43.0% (P = 0.02), deaths from coronary heart disease (CHD) by 30.0% (P < 0.01), and all-cause mortality by 35.8% (P = 0.001). CONCLUSIONS: This study, for the first time, demonstrated that long-term Xuezhikang therapy resulted in significant reduction in cardiovascular events and death in Chinese hypertensive patients with previous MI in a safe manner.


Subject(s)
Coronary Artery Disease/prevention & control , Drugs, Chinese Herbal/therapeutic use , Hypertension/diet therapy , Myocardial Infarction/complications , Adolescent , Adult , Aged , China/epidemiology , Coronary Artery Disease/etiology , Double-Blind Method , Female , Humans , Hypertension/complications , Hypertension/mortality , Incidence , Kaplan-Meier Estimate , Lipoproteins/blood , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL