Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Sci Data ; 10(1): 839, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38030700

ABSTRACT

Livestock rearing is a major source of livelihood for food and income in dryland Asia. Increasing livestock density (LSKD) affects ecosystem structure and function, amplifies the effects of climate change, and facilitates disease transmission. Significant knowledge and data gaps regarding their density, spatial distribution, and changes over time exist but have not been explored beyond the county level. This is especially true regarding the unavailability of high-resolution gridded livestock data. Hence, we developed a gridded LSKD database of horses and small ruminants (i.e., sheep & goats) at high-resolution (1 km) for Kazakhstan (KZ) from 2000-2019 using vegetation proxies, climatic, socioeconomic, topographic, and proximity forcing variables through a random forest (RF) regression modeling. We found high-density livestock hotspots in the south-central and southeastern regions, whereas medium-density clusters in the northern and northwestern regions of KZ. Interestingly, population density, proximity to settlements, nighttime lights, and temperature contributed to the efficient downscaling of district-level censuses to gridded estimates. This database will benefit stakeholders, the research community, land managers, and policymakers at regional and national levels.


Subject(s)
Ecosystem , Livestock , Animals , Goats , Horses , Kazakhstan , Sheep
2.
Front Pharmacol ; 14: 1137783, 2023.
Article in English | MEDLINE | ID: mdl-36937838

ABSTRACT

We investigated the efficacy of a small molecule ASR-600, an analog of Urolithin A (Uro A), on blocking androgen receptor (AR) and its splice variant AR-variant 7 (AR-V7) signaling in castration-resistant prostate cancer (CRPC). ASR-600 effectively suppressed the growth of AR+ CRPC cells by inhibiting AR and AR-V7 expressions; no effect was seen in AR- CRPC and normal prostate epithelial cells. Biomolecular interaction assays revealed ASR-600 binds to the N-terminal domain of AR, which was further confirmed by immunoblot and subcellular localization studies. Molecular studies suggested that ASR-600 promotes the ubiquitination of AR and AR-V7 resulting in the inhibition of AR signaling. Microsomal and plasma stability studies suggest that ASR-600 is stable, and its oral administration inhibits tumor growth in CRPC xenografted castrated and non-castrated mice. In conclusion, our data suggest that ASR-600 enhances AR ubiquitination in both AR+ and AR-V7 CRPC cells and inhibits their growth in vitro and in vivo models.

3.
Transl Res ; 231: 92-101, 2021 05.
Article in English | MEDLINE | ID: mdl-33279680

ABSTRACT

Currently, clinicians rely on clinical nomograms to stratify progression risk at the time of diagnosis in patients with prostate cancer (CaP). However, these tools may not accurately distinguish aggressive potential in low-grade CaP. The current study determined the diagnostic potential of 3 molecular markers (ROCK1, RUNX3, and miR-301a) in terms of their ability to identify which low-grade tumors are likely to progress. Real-time PCR and immunohistochemical analysis were used to assess ROCK1, RUNX3, and miR-301a expression profiles in 118 serum and needle biopsy specimens. Expressions of ROCK1 and miR-301a were found to be significantly higher in Gleason 6 and 7 CaP as compared to BPH, while an inverse trend was observed with RUNX3. Further, incorporation of all 3 molecular markers significantly improved clinical nomograms' diagnostic accuracy and correlated with disease progression. Hence, in conclusion, the inclusion of these 3 molecular markers identified aggressive phenotype and predicted disease progression in low-grade CaP tumors at the time of diagnosis.


Subject(s)
Biomarkers, Tumor/blood , Core Binding Factor Alpha 3 Subunit/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , rho-Associated Kinases/metabolism , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Biopsy , Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Middle Aged , Polymerase Chain Reaction , rho-Associated Kinases/genetics
4.
Mol Cancer Ther ; 19(12): 2422-2431, 2020 12.
Article in English | MEDLINE | ID: mdl-33087513

ABSTRACT

Notch1 activation triggers significant oncogenic signaling that manifests as enhanced metastatic potential and tumorigenesis in colorectal cancer. Novel small-molecule inhibitors, mainly plant-derived analogs, have low toxicity profiles and higher bioavailability. In this study, we have developed a small molecule, ASR490, by modifying structure of naturally occurring compound Withaferin A. ASR490 showed a growth-inhibitory potential by downregulating Notch1 signaling in HCT116 and SW620 cell lines. Docking studies and thermal shift assays confirmed that ASR490 binds to Notch1, whereas no changes in Notch2 and Notch3 expression were seen in colorectal cancer cells. Notch1 governs epithelial-to-mesenchymal transition signaling and is responsible for metastasis, which was abolished by ASR490 treatment. To further confirm the therapeutic potential of ASR490, we stably overexpressed Notch1 in HCT-116 cells and determined its inhibitory potential in transfected colorectal cancer (Notch1/HCT116) cells. ASR490 effectively prevented cell growth in both the vector (P = 0.005) and Notch1 (P = 0.05) transfectants. The downregulation of Notch1 signaling was evident, which corresponded with downregulation of mesenchymal markers, including N-cadherin and ß-catenin and induction of E-cadherin in HCT-116 transfectants. Intraperitoneal administration of a 1% MTD dose of ASR490 (5 mg/kg) effectively suppressed the tumor growth in control (pCMV/HCT116) and Notch1/HCT116 in xenotransplanted mice. In addition, downregulation of Notch1 and survival signaling in ASR-treated tumors confirmed the in vitro results. In conclusion, ASR490 appears to be a potent agent that can inhibit Notch1 signaling in colorectal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Receptor, Notch1/genetics , Biomarkers , Cell Line, Tumor , HCT116 Cells , Humans , Receptor, Notch1/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism
5.
Oncol Lett ; 20(1): 850-860, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32566012

ABSTRACT

Due to a lack of mechanistic insights, muscle-invasive bladder cancer (MIBC) remains incurable and is one of the most lethal types of cancer in the United States. The present study investigated changes in the molecular signatures of MIBC cells (TCCSUP and HT1376) after treatment with a novel small molecule, ASR488, to gain knowledge of the mechanisms that inhibited MIBC cell growth. ASR488 treatment initiated apoptotic signaling in MIBC cells. Pathway enrichment analysis was used to analyze the changes in function of differentially expressed genes. Gene Ontology analysis, as well as Kyoto Encyclopedia of Genes and Genomes analysis, was also performed. These analyses along with reactome pathway enrichment analyses indicated that the genes upregulated in the ASR488-treated cells are involved in focal adhesion, neurotrophin signaling, p53 signaling, endoplasmic reticulum functioning in terms of protein processing, and pathways related to bladder cancer. The genes downregulated in ASR488-treated MIBC cells were mainly involved in DNA replication, mismatch repair, RNA degradation, nucleotide excision repair and TGFß signaling (P<0.05). Furthermore, reverse transcription-quantitative PCR analysis revealed an increase in transcripts of the most upregulated genes in ASR 488-treated MIBC cells: CPEB1 (36-fold), IL11 (30-fold), SFN (20.12-fold) and CYP4F11 (15.8-fold). In conclusion, the analysis of biological functions of the most differentially expressed genes revealed possible mechanisms that may be associated with the aggressiveness of MIBC.

6.
Oncogenesis ; 9(2): 23, 2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32066655

ABSTRACT

Epidemiological evidence suggests that cadmium (Cd) is one of the causative factors of prostate cancer, but the effect of Cd on benign prostatic hyperplasia (BPH) remains unclear. This study aimed to determine whether Cd exposure could malignantly transform BPH1 cells and, if so, to dissect the mechanism of action. We deciphered the molecular signaling responsible for BPH1 transformation via RNA-sequencing and determined that Cd induced the expression of zinc finger of the cerebellum 2 (ZIC2) in BPH1 cells. We noted Cd exposure increased ZIC2 expression in the Cd-transformed BPH1 cells that in turn promoted anchorage-independent spheroids and increased expression of stem cell drivers, indicating their role in stem cell renewal. Subsequent silencing of ZIC2 expression in transformed cells inhibited spheroid formation, stem cell marker expression, and tumor growth in nude mice. At the molecular level, ZIC2 interacts with the glioma-associated oncogene family (GLI) zinc finger 1 (GLI1), which activates prosurvival factors (nuclear factor NFκB, B-cell lymphoma-2 (Bcl2), as well as an X-linked inhibitor of apoptosis protein (XIAP)) signaling in Cd-exposed BPH1 cells. Conversely, overexpression of ZIC2 in BPH1 cells caused spheroid formation confirming the oncogenic function of ZIC2. ZIC2 activation and GLI1 signaling induction by Cd exposure in primary BPH cells confirmed the clinical significance of this oncogenic function. Finally, human BPH specimens had increased ZIC2 versus adjacent healthy tissues. Thus, we report direct evidence that Cd exposure induces malignant transformation of BPH via activation of ZIC2 and GLI1 signaling.

7.
Toxicol Appl Pharmacol ; 375: 57-63, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31082426

ABSTRACT

The aim of the present study was to investigate the genetic signatures of cadmium-transformed prostate epithelial (CTPE) cells and to identify the potential molecular signaling involved in their malignant transformation. The dataset contained normal prostate epithelial (RWPE-1) and CTPE cells. To further examine the biological functions of the identified differentially expressed genes (DEGs), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway enrichment analyses were performed. In total, 2357 DEGs were identified, including 1083 upregulated genes and 1274 downregulated genes. GO, KEGG, and Reactome pathway enrichment analyses indicated that upregulated genes were significantly enriched in ECM-receptor, focal adhesion, TGFß signaling, and syndecan interactions, while downregulated genes were mainly involved in cell cycle regulation, arachidonic acid metabolism, oxidative phosphorylation, and folate biosynthesis (p < .05). The top upregulated (SATB1 (p < .0001), EYA2 (p < .0001) and KPNA7 (p < .0027)) and downregulated (PITX2 (p < .0007), PDLIM4 (p < .0020) and FABP5 (p < .0007)) genes were further validated via qRT-PCR analysis. In conclusion, the present study profiled DEGs in RWPE-1 and CTPE cells and identified gene pathways that may be associated with malignant transformation and tumor progression.


Subject(s)
Cadmium/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Prostate/cytology , Prostatic Neoplasms/chemically induced , Cell Line, Tumor , Cluster Analysis , Gene Regulatory Networks , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/metabolism
8.
Urol Oncol ; 37(7): 492-502, 2019 07.
Article in English | MEDLINE | ID: mdl-31006613

ABSTRACT

PURPOSE: The role of androgen receptor (AR) signaling in bladder cancer (BCa) is not fully characterized. This study aimed to delineate the role of AR signaling in BCa and to determine whether the combination of AR inhibitor, Enzalutamide (Enz), and Cisplatin (Cis) efficiently inhibit the growth of BCa cells. METHODS: AR expression was determined in 89 human urothelial BCa specimens by immunohistochemistry. A panel of BCa cell lines was treated with Cis, Enz, or a combination of both (Enz + Cis). We determined the expression of AR, changes in apoptotic signaling, DNA damage, and analyzed effect on epithelial mesenchymal transformation markers. RESULT: AR expression was detected in 61.4% of tumors from male BCa patients. Inhibition of AR signaling by Enz effectively inhibited the growth of AR+ BCa cells by inducing apoptosis (26%) in AR+ TCCSUP (P = 0.0201) and J82 (15%, P = 0.0386) cells. Interestingly, Enz + Cis synergistically inhibited the proliferation of BCa cells even at low concentrations by inducing proapoptotic signaling in AR+ BCa cells. Invasive and migratory potential of TCCSUP and J82 cells were reduced with Enz + Cis treatment, and associated with down-regulation of mesenchymal markers. CONCLUSIONS: A high percentage of the bladder tumors from male patients in our cohort expressed AR. The combination of Enz and Cis synergistically inhibited growth of BCa cells more efficiently than single agent alone. This supports the rationale for future investigation of AR antagonists in combination with standard chemotherapy in MIBC.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Cisplatin/therapeutic use , Receptors, Androgen/metabolism , Urinary Bladder Neoplasms/drug therapy , Androgen Receptor Antagonists/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Benzamides , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cohort Studies , DNA Damage/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Male , Middle Aged , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Signal Transduction/drug effects , Treatment Outcome , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
9.
Toxicol Appl Pharmacol ; 373: 62-68, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31002860

ABSTRACT

Earlier, we reported that chronic cadmium (Cd)-exposure to prostate epithelial (RWPE-1) cells causes defective autophagy, which leads to the transformation of a malignant phenotype in both in vitro and in vivo models. However, the upstream events responsible for defective autophagy are yet to be delineated. The present study suggests that chronic Cd exposure induces endoplasmic reticulum (ER) stress that triggers the phosphorylation of stress transducers [protein kinase R-like ER Kinase- (PERK), eukaryotic translation initiation factor 2-alpha- (eIF2-α) and Activating Transcription Factor 4 -(ATF-4)], resulting in defective autophagy that protects Cd-exposed RWPE-1 cells. On the other hand, inhibition of the ATF4 stress inducer by siRNA blocked the Cd-induced defective autophagy in transforming cells. While dissecting the upstream activators of ER stress, we found that increased expression of reactive oxygen species (ROS) is responsible for ER stress in Cd-exposed RWPE-1 cells. Overexpression of antioxidants (SOD1/SOD2) mitigates Cd-induced ROS that results in inhibition of ER stress and autophagy in prostate epithelial cells. These results suggest that the induction of ROS and subsequent ER stress are responsible for defective autophagy in Cd-induced transformation in prostate epithelial cells.


Subject(s)
Autophagy/drug effects , Cadmium/toxicity , Cell Transformation, Neoplastic/chemically induced , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Prostate/drug effects , Prostatic Neoplasms/chemically induced , Autophagy-Related Proteins/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Oxidative Stress/drug effects , Phosphorylation , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
10.
Urol Oncol ; 36(11): 503.e9-503.e15, 2018 11.
Article in English | MEDLINE | ID: mdl-30195463

ABSTRACT

BACKGROUND: Prostate-specific antigen screening for prostate cancer (CaP) remains controversial. This study establishes the role of microRNA 301a (miR-301a) as a supplemental biomarker that can distinguish between patients with benign prostate hyperplasia and clinically significant CaP. We evaluate the ability of miR-301a to predict the adverse pathology of CaP. METHODS: In the first cohort, serum and prostate tumor samples were obtained from thirteen patients with Benign prostate hyperplasia (BPH), twelve patients with Gleason 6, and sixteen patients with Gleason 7 prostate adenocarcinoma. In the second cohort, 40 prostatectomy cases were selected (BPH:12, Gleason 6:12 and Gleason 7:16). MiRNA was extracted from serum and tumor samples. Quantitative reverse transcription-polymerase chain reaction was performed for detection of miR-301a. To understand the molecular role of miR-301a, we performed cell viability, Western blots, promoter analysis, overexpression, and silencing studies in BPH and DU-145 cell lines. RESULTS: MiR-301a demonstrated a significantly higher expression in both serum and tumor tissue in patients with CaP when compared to patients with BPH (P = 0.011 and 0.013 for serum and tissue expression, respectively). Expression of miR-301a in prostatectomy specimens correlated with increased Gleason score. We demonstrated that miR-301a inhibited the pro-apoptotic function of RUNX3, and activated ROCK1-mediated pro-survival signal in CaP. Silencing miR-301a initiated the pro-apoptotic function of RUNX3 by inhibiting ROCK1 expression in CaP cells. CONCLUSIONS: Expression of miR-301a could be a valuable adjunct tool for stratifying patients with elevated prostate-specific antigen, as well as those diagnosed with CaP. Including the miR-301a as an additional variable in MSKCC post-prostatectomy nomogram improved its ability in facilitating clinical decision-making.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/genetics , MicroRNAs/biosynthesis , Prostatic Neoplasms/diagnosis , Adenocarcinoma/mortality , Adult , Aged , Area Under Curve , Humans , Male , MicroRNAs/analysis , Middle Aged , Nomograms , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , ROC Curve
11.
Carcinogenesis ; 39(12): 1537-1547, 2018 12 31.
Article in English | MEDLINE | ID: mdl-30124785

ABSTRACT

Chemopreventive effects and associated mechanisms of withaferin A (WA) against intestinal and colon carcinogenesis remain unknown. We investigated the chemopreventive effect of WA on transgenic adenomatous polyposis coli (APCMin/+) mouse and chemically induced azoxymethane/dextran sodium sulfate (AOM/DSS) models of intestinal and colon carcinogenesis. Oral WA administration (4 and 3 mg/kg) inhibited tumor initiation and progression of intestinal polyps formation in APCMin/+ mice and colon carcinogenesis in the AOM/DSS mouse model. WA-administered mice showed a significant reduction in both number [duodenum, 33% (P > 0.05); jejunum, 32% (P < 0.025); ileum, 43% ( P < 0.001); and colon 59% (P < 0.01] and size of polyps in APCMin/+ mice compared with the respective controls. Similarly, tumor multiplicity was significantly reduced (P < 0.05) in the colon of WA-administered AOM/DSS mice. Pathological analysis showed reduced adenomas and tissue inflammation in WA-administered mouse models. Molecular studies suggested that WA inhibited the expression of inflammatory (interluekin-6, tumor necrosis factor-alpha and cyclooxygenase-2), pro-survival (pAKT, Notch1 and NF-κB) markers in APCMin/+ and AOM/DSS models. The results suggest that WA is a potent agent for preventing colon carcinogenesis and further investigation is required to show clinical utility of the agent.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinogenesis/drug effects , Colon/drug effects , Colonic Neoplasms/prevention & control , Inflammation/drug therapy , Withanolides/pharmacology , Animals , Chemoprevention/methods , Colon/pathology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Mol Carcinog ; 57(10): 1332-1341, 2018 10.
Article in English | MEDLINE | ID: mdl-30069922

ABSTRACT

Androgen ablation therapy is the primary therapeutic option for locally advanced and metastatic castration-resistant prostate cancer (CRPC). We investigated therapeutic effect of a dietary metabolite Urolithin A (UroA) and dissected the molecular mechanism in CRPC cells. Treatment with UroA inhibited cell proliferation in both androgen receptor-positive (AR+ ) (C4-2B) and androgen receptor-negative (AR- ) (PC-3) cells however, AR+ CaP cells were more sensitive to UroA treatment as compared with AR- CaP cells. Inhibition of the AR signaling was responsible for the UroA effect on AR+ CaP cells. Ectopic expression of AR in PC-3 cells sensitized them to UroA treatment as compared to the vector-expresseing PC-3 cells, which suggests that AR could be a target of UroA. Similarly, in enzalutamide-resistant C4-2B cells, a downregulation of AR expression also suppressed cell proliferation which was observed with the UroA treatment. Oral administration of UroA significantly suppressed the growth of C4-2B xenografts (P = 0.05) compared with PC-3 xenografts (P = 0.069) without causing toxicity to animals. Immunohistochemistry analysis confirmed in vitro findings such as downregulation of AR/pAKT signaling in UroA-treated C4-2B tumors, which suggests that UroA may be a potent chemo-preventive and therapeutic agent for CRPC.


Subject(s)
Cell Proliferation/drug effects , Coumarins/pharmacology , Down-Regulation/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Burden/drug effects , Tumor Burden/genetics
13.
Cancer Lett ; 408: 121-129, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28844710

ABSTRACT

Chronic exposure to cadmium is known to be a risk factor for human prostate cancer. Despite over-whelming evidence of cadmium causing carcinogenicity in humans, the specific underlying molecular mechanisms that govern metal-induced cellular transformation remain unclear. Acute exposure (up to 72 h) to cadmium induces apoptosis in normal prostate epithelial cells (RWPE-1), while chronic exposure (>1 year) transforms these cells to a malignant phenotype (cadmium-transformed prostate epithelial cells; CTPE). Increased expression of autophagy-regulated genes; Plac8, LC3B and Lamp-1; in CTPE cells was associated with cadmium-induced transformation. Increased expression of Plac8, a regulator of autophagosome/autolysosome fusion, facilitates the pro-survival function of autophagy and upregulation of pAKT(ser473) and NF-κß, to allow CTPE to proliferate. Likewise, inhibition of Plac8 suppresses CTPE cell growth. Additionally, overexpression of Plac8 in RWPE-1 cells induces resistance to cadmium toxicity. Pharmacological inhibitors and an inducer of autophagy failed to affect Plac8 expression and CTPE cell viability, suggesting a unique role for Plac8 in cadmium-induced prostate epithelial cell transformation. These results support a role for Plac8 as an essential component in the cadmium-induced transformation of normal prostate epithelial cells to a cancerous state.


Subject(s)
Autophagy/drug effects , Cadmium/toxicity , Cell Transformation, Neoplastic/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proteins/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Br J Cancer ; 117(1): 56-64, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28588318

ABSTRACT

BACKGROUND: Cadmium, an established carcinogen, is a risk factor for prostate cancer. Induction of autophagy is a prerequisite for cadmium-induced transformation and metastasis. The ability of Psoralidin (Pso), a non-toxic, orally bioavailable compound to inhibit cadmium-induced autophagy to prevent prostate cancer was investigated. METHODS: Psoralidin was studied using cadmium-transformed prostate epithelial cells (CTPE), which exhibit high proliferative, invasive and colony forming abilities. Gene and protein expression were evaluated by qPCR, western blot, immunohistochemistry and immunofluorescence. Xenograft models were used to study the chemopreventive effects in vivo. RESULTS: Cadmium-transformed prostate epithelial cells were treated with Pso resulting in growth inhibition, without causing toxicity to normal prostate epithelial cells (RWPE-1). Psoralidin-treatment of CTPE cells inhibited the expression of Placenta Specific 8, a lysosomal protein essential for autophagosome and autolysosome fusion, which resulted in growth inhibition. Additionally, Pso treatment caused decreased expression of pro-survival signalling proteins, NFκB and Bcl2, and increased expression of apoptotic genes. In vivo, Pso effectively suppressed CTPE xenografts growth, without any observable toxicity. Tumours from Pso-treated animals showed decreased autophagic morphology, mesenchymal markers expression and increased epithelial protein expression. CONCLUSIONS: These results confirm that inhibition of autophagy by Pso plays an important role in the chemoprevention of cadmium-induced prostate carcinogenesis.


Subject(s)
Autophagy/drug effects , Benzofurans/pharmacology , Cadmium/adverse effects , Carcinogenesis/drug effects , Coumarins/pharmacology , Prostate/drug effects , Prostatic Neoplasms/chemically induced , Animals , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Blotting, Western , Carcinogenesis/chemically induced , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice, Nude , NF-kappa B/drug effects , NF-kappa B/metabolism , Neoplasm Transplantation , Polymerase Chain Reaction , Proteins/drug effects , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
15.
Mol Carcinog ; 56(3): 1127-1136, 2017 03.
Article in English | MEDLINE | ID: mdl-27753148

ABSTRACT

We have previously reported that high aldehyde dehydrogenase (ALDH) enzyme activity in breast cancer cells results in breast cancer stem cell (BCSC) properties by upregualting Notch-1 and epithelial mesenchymal markers. This results in chemoresistance in breast cancer. Here, we examined the functional and clinical significance of ALDH expression by measuring the ALDH levels in breast cancer tissues by immunohistochemistry. There was a significantly higher ALDH expression in higher grade breast cancer tumor tissues (Grade- II and III) versus normal breast tissues. Injection of BCSC (ALDH+ and CD44+ /CD22- ) cells resulted in aggressive tumor growth in athymic mice versus ALDH- cells. The ALDH+ and CD44+ /CD22- tumors grow rapidly and are larger than ALDH- tumors which were slow growing and smaller. Molecularly, ALDH+ tumors expressed higher expression of Notch-1 and EMT markers than ALDH- tumors. Oral administration of the naturally occurring Psoralidin (Pso, 25 mg/kg of body weight) significantly inhibited the growth in ALDH+ and ALDH- tumors as well. Psoralidin inhibited Notch-1 mediated EMT activation in ALDH+ and ALDH- tumors-this confirms our in vitro findings. Our results suggest that Notch-1 could be an attractive target and inhibition of Notch-1 by Psoralidin may prevent pathogenesis of breast cancer as well as metastasis. © 2016 Wiley Periodicals, Inc.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Benzofurans/administration & dosage , Breast Neoplasms/drug therapy , Coumarins/administration & dosage , Neoplastic Stem Cells/drug effects , Receptor, Notch1/metabolism , Animals , Benzofurans/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Coumarins/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
16.
Oncotarget ; 7(33): 53751-53761, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27447565

ABSTRACT

We previously reported that withaferin A (WA), a natural compound, deters prostate cancer by inhibiting AKT while inducing apoptosis. In the current study, we examined its chemopreventive efficacy against carcinogenesis in the prostate using the transgenic adenocarcinoma of mouse prostate (TRAMP) model. Two distinct sets of experiments were conducted. To determine whether WA delays tumor progression, it was given before cancer onset, at week 6, and until week 44. To determine its effect after the onset of prostate cancer, it was given from weeks 12 to 35. In both strategies, oral administration of WA effectively suppressed tumor burden when compared to vehicle-treated animals. No toxicity was seen in treated animals at gross pathological examination. Western blot analysis and immunohistochemistry of tumor sections revealed that in TRAMP controls, AKT and pAKT were highly expressed while nuclear FOXO3a and Par-4 were downregulated. On the contrary, treated mice showed inhibition of AKT signaling and activation of FOX03a-Par-4-induced cell death. They also displayed inhibition of mesenchymal markers such as ß-catenin, vimentin, and snail as well as upregulation of E-cadherin. Because expressions of the angiogenic markers factor VIII and retic were downregulated, an anti-angiogenic role of WA is suggested. Overall, our results suggest that WA could be a promising anti-cancer agent that effectively inhibits carcinogenesis of the prostate.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Prostatic Neoplasms/pathology , Withanolides/pharmacology , Administration, Oral , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Urol Oncol ; 34(8): 336.e13-20, 2016 08.
Article in English | MEDLINE | ID: mdl-27133223

ABSTRACT

PURPOSE: The diagnosis and treatment of prostate cancer (CaP) continues to be challenging, as prostate-specific antigen (PSA) appears to be overly sensitive and biopsy is the only reliable method for confirmation. Hence, the goal of the study is to identify a biomarker that could distinguish malignant cancer from benign prostatic hyperplasia (BPH) during the early diagnosis of the disease. MATERIALS AND METHODS: A total of 75 formalin fixed paraffin embedded (FFPE) with matching controls, 4 paired metastatic tumors, 6 fresh tumor tissues and BPH (13 cases) with their clinical diagnosis were selected for this study. Prostate cancer cell lines and normal prostate epithelial cell lines were obtained from ATCC and subjected to phenotypic analysis. RESULTS: We observed significant differential expression of miR-301a in CaP samples in comparison to BPH and adjacent benign samples. The overexpression of miR-301a activates the invasion/migration of CaP cells. In contrast, silencing miR-301a expression inhibited the colony-forming ability, adhesion, invasion and migration of CaP cells. Similarly, the overexpression of miR-301a increased cell motility in normal RWPE-1 prostate epithelial cells. Our results suggest that miR-301a is differentially expressed between BPH and CaP specimens and that the expression of miR-301a correlates with biochemical recurrence and/or metastasis in CaP patients. CONCLUSIONS: The expression of miR-301a could be a potential marker for metastasis in CaP patients. Detecting miR-301a expression during diagnosis will avoid wait and watch timelines, thus preventing morbidity.


Subject(s)
Biomarkers, Tumor/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/diagnosis , Cell Line, Tumor , Cell Movement , Humans , Male , Prognosis , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism
18.
Hepatology ; 59(5): 1886-99, 2014 May.
Article in English | MEDLINE | ID: mdl-24285179

ABSTRACT

UNLABELLED: Mouse Double Minute homolog 4 (MDM4) gene up-regulation often occurs in human hepatocellular carcinoma (HCC), but the molecular mechanisms responsible for its induction remain poorly understood. Here we investigated the role of the phosphoinositide-3-kinase/v-akt murine thymoma viral oncogene homolog/mammalian target of rapamycin (PI3K/AKT/mTOR) axis in the regulation of MDM4 levels in HCC. The activity of MDM4 and the PI3K/AKT/mTOR pathway was modulated in human HCC cell lines by way of silencing and overexpression experiments. Expression of main pathway components was analyzed in an AKT mouse model and human HCCs. MDM4 inhibition resulted in growth restraint of HCC cell lines both in vitro and in vivo. Inhibition of the PI3K-AKT and/or mTOR pathways lowered MDM4 protein levels in HCC cells and reactivated p53-dependent transcription. Deubiquitination by ubiquitin-specific protease 2a and AKT-mediated phosphorylation protected MDM4 from proteasomal degradation and increased its protein stability. The eukaryotic elongation factor 1A2 (EEF1A2) was identified as an upstream inducer of PI3K supporting MDM4 stabilization. Also, we detected MDM4 protein up-regulation in an AKT mouse model and a strong correlation between the expression of EEF1A2, activated/phosphorylated AKT, and MDM4 in human HCC (each rho > 0.8, P < 0.001). Noticeably, a strong activation of this cascade was associated with shorter patient survival. CONCLUSION: The EEF1A2/PI3K/AKT/mTOR axis promotes the protumorigenic stabilization of the MDM4 protooncogene in human HCC by way of a posttranscriptional mechanism. The activation level of the EEF1A2/PI3K/AKT/mTOR/MDM4 axis significantly influences the survival probability of HCC patients in vivo and may thus represent a promising molecular target.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/physiology , Peptide Elongation Factor 1/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , Aged , Animals , Carcinoma, Hepatocellular/mortality , Cell Cycle Proteins , Female , Hep G2 Cells , Humans , Liver Neoplasms/mortality , Male , Mice , Middle Aged , Signal Transduction/physiology
19.
PLoS One ; 7(2): e31430, 2012.
Article in English | MEDLINE | ID: mdl-22384021

ABSTRACT

Dupuytren's disease (DD) is a benign, fibroproliferative disease of the palmar fascia, with excessive extracellular matrix (ECM) deposition and over-production of cytokines and growth factors, resulting in digital fixed flexion contractures limiting hand function and patient quality of life. Surgical fasciectomy is the gold standard treatment but is invasive and has associated morbidity without limiting disease recurrence. Injectable Collagenase Clostridium histolyticum (CCH)--Xiaflex®--is a novel, nonsurgical option with clinically proven in vivo reduction of DD contractures but with limited in vitro data demonstrating its cellular and molecular effects. The aim of this study was to delineate the effects of CCH on primary fibroblasts isolated from DD and non-DD anatomical sites (using RTCA, LDH, WST-1, FACS, qRT-PCR, ELISA and In-Cell Quantitative Western Blotting) to compare the efficacy of varying concentrations of Xiaflex® against a reagent grade Collagenase, Collagenase A. Results demonstrated that DD nodule and cord fibroblasts had greater proliferation than those from fat and skin. Xiaflex® exposure resulted in dose- and time-dependent inhibition of cellular spreading, attachment and proliferation, with cellular recovery after enzyme removal. Unlike Collagenase A, Xiaflex® did not cause apoptosis. Collagen expression patterns were significantly (p<0.05) different in DD fibroblasts across anatomical sites - the highest levels of collagen I and III were detected in DD nodule, with DD cord and fat fibroblasts demonstrating a smaller increase in both collagen expression relative to DD skin. Xiaflex® significantly (p<0.05) down-regulated ECM components, cytokines and growth factors in a dose-dependent manner. An in vitro scratch wound assay model demonstrated that, at low concentrations, Xiaflex® enabled a faster fibroblast reparatory migration into the wound, whereas, at high concentrations, this process was significantly (p<0.05) inhibited. This is the first report elucidating potential mechanisms of action of Xiaflex® on Dupuytren fibroblasts, offering a greater insight and a better understanding of its effect in DD.


Subject(s)
Dupuytren Contracture/drug therapy , Dupuytren Contracture/metabolism , Fibroblasts/drug effects , Gene Expression Regulation , Microbial Collagenase/metabolism , Microbial Collagenase/pharmacology , Adult , Aged , Aged, 80 and over , Apoptosis , Cell Survival , Dose-Response Relationship, Drug , Female , Fibroblasts/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Necrosis , Time Factors , Treatment Outcome
20.
Stem Cells Dev ; 21(4): 609-22, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21612554

ABSTRACT

Dupuytren's disease (DD) is a fibroproliferative disorder characterized by aberrant proliferation of myofibroblasts, the source of which remains unknown. Recent studies indicate that circulating and tissue-resident mesenchymal stem cells (MSCs) can differentiate into myofibroblasts. Therefore, the aim of this study was to profile MSCs from phenotypically distinct DD sites including cord, nodule, skin overlying nodule (SON), and perinodular fat (PNF) compared with unaffected internal controls, that is, distant palmar fat (DPF) and transverse palmar fascia (Skoog's fibers) as well as external control carpal tunnel (CT) tissue including skin, fat, and fascia. Freshly isolated primary fibroblasts as well as cells grown up to passage 5 (P5) from DD (n=27) and CT (n=14) samples were analyzed for the presence of established MSC markers CD73, CD90, and CD105 and absence of hematopoietic marker CD34 using fluorescence-activated cell sorting, in-cell quantitative western blotting, immunohistochemistry, and immunocytochemistry. Freshly isolated cells from SON, PNF, and cord biopsies had a higher number of CD34(-)73(+)90(+)105(+) cells compared with Skoog's fibers and CT controls. P3 cells obtained from all DD biopsies compared with CT samples differentiated into osteocytes, adipocytes, and chondrocytes. P3 cord and nodule cells expressed intense α-smooth muscle actin staining compared with skin and fat cells. Stem cell markers including stem cell factor, MSC-homing marker CXCR4, and Wnt/ß-catenin downregulator Dkk-1 were all upregulated in SON and PNF compared with CT skin and CT fat, respectively, as shown by real-time quantitative polymerase chain reaction. However, osteogenic marker OSF-1 had a significantly higher expression in the PNF (P=0.002) and cord (P=0.01) compared with the nodule. In conclusion, we have shown the presence of MSCs in specific DD tissue phenotypes compared with internal and external control tissue. These findings provide preliminary support for a potential alternative source of disease myofibroblasts originating from sites such as SON and PNF as opposed to palmar fascia alone.


Subject(s)
Adipose Tissue , Cell Differentiation , Dupuytren Contracture , Mesenchymal Stem Cells , Myofibroblasts , Skin , Adipose Tissue/metabolism , Adipose Tissue/pathology , Antigens, Differentiation/metabolism , Cells, Cultured , Dupuytren Contracture/metabolism , Dupuytren Contracture/pathology , Dupuytren Contracture/physiopathology , Dupuytren Contracture/therapy , Hand/pathology , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Skin/metabolism , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...