Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Front Med (Lausanne) ; 10: 1070362, 2023.
Article in English | MEDLINE | ID: mdl-36936230

ABSTRACT

Background: Peritoneal metastasis (PM) from colorectal cancer carries a dismal prognosis despite extensive cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC). With a median time to recurrence of 11-12 months, there is a need for novel therapies. Radspherin® consists of the α-emitting radionuclide radium-224 (224Ra), which has a half-life of 3.6 days and is adsorbed to a suspension of biodegradable calcium carbonate microparticles that are designed to give short-range radiation to the serosal peritoneal surface linings, killing free-floating and/or tumor cell clusters that remain after CRS-HIPEC. Methods: A first-in-human phase 1 study (EudraCT 2018-002803-33) was conducted at two specialized CRS-HIPEC centers. Radspherin® was administered intraperitoneally 2 days after CRS-HIPEC. Dose escalation at increasing activity dose levels of 1-2-4-7-MBq, a split-dose repeated injection, and expansion cohorts were used to evaluate the safety and tolerability of Radspherin®. The aim was to explore the recommended dose and biodistribution using gamma-camera imaging. The results from the planned safety interim analysis after the completion of the dose-limiting toxicity (DLT) period of 30 days are presented. Results: Twenty-three patients were enrolled: 14 in the dose escalation cohort, three in the repeated cohort, and six in the expansion cohort. Of the 23 enrolled patients, seven were men and 16 were women with a median age of 64 years (28-78). Twelve patients had synchronous PM stage IV and 11 patients had metachronous PM [primary stage II; (6) and stage III; (5)], with a disease-free interval of 15 months (3-30). The peritoneal cancer index was median 7 (3-19), operation time was 395 min (194-515), and hospital stay was 12 days (7-37). A total of 68 grade 2 adverse events were reported for 17 patients during the first 30 days; most were considered related to CRS and/or HIPEC. Only six of the TEAEs were evaluated as related to Radspherin®. One TEAE, anastomotic leakage, was reported as grade 3. Accordion ≥3 grade events occurred in a total of four of the 23 patients: reoperation due to anastomotic leaks (two) and drained abscesses (two). No DLT was documented at the 7 MBq dose level that was then defined as the recommended dose. The biodistribution of Radspherin® showed a relatively even peritoneal distribution. Conclusion: All dose levels of Radspherin® were well tolerated, and DLT was not reached. No deaths occurred, and no serious adverse events were considered related to Radspherin®.Clinical Trial Registration: Clinicaltrials.gov, NCT03732781.

2.
Cell Death Dis ; 11(10): 868, 2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33067425

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Pathol Res Pract ; 215(11): 152638, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31551175

ABSTRACT

AIM: Long non-coding RNA (lncRNA) is currently considered to play an important regulatory role in various diseases, including tumors, at present a hot topic in research. As a non-coding transcription product of imprinted gene, LncRNA H19 is expressed as a parent imprinted maternal allele without protein-coding ability. Increasing evidence indicates that LncH19 may be a new tumor marker for early clinical diagnosis and prognosis judgment. In this study, LncH19 expression was investigated by RNA in situ hybridization for further exploring the clinicopathological role of its expression in esophageal squamous cell cancer (ESCC). METHODS: 121 tumor samples and seven cases of adjacent non-tumor tissues from esophageal cancer patients were detected by RNA in situ hybridization (ISH) and the ISH staining was graded with modified Allred scoring. RESULTS: While no LncH19 expression in the tumor adjacent to normal epithelia was disclosed with the technology, significantly higher levels of LncH19 expression were detected in the tumors obtained from the patients who died within one year after surgery, compared to the expression in those tumors from the patients who survived longer than five years after the same treatment regimen (P = 0.001). In addition, LncH19 expression was verified to correlate with a larger tumor size (P = 0.002) and a higher UICC stage (P = 0.001). CONCLUSION: Our LncH19 ISH data verify the involvement of LncH19 in ESCC. Higher levels of LncH19 expression were not only detected in tumors with larger size and in clinical late stage, but also significantly associated with shorter survival, strongly indicating its clinical significance in the malignant progression of ESCC and useful value as a poor prognostic factor for the patients.


Subject(s)
Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , RNA, Long Noncoding/biosynthesis , Adult , Aged , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis
4.
Front Oncol ; 9: 1494, 2019.
Article in English | MEDLINE | ID: mdl-31998647

ABSTRACT

The Guanylate binding proteins (GBPs) are a family of large GTPases and the most studied GBP family member is the guanylate binding protein 1 (GBP1). Earlier studies revealed that GBP1 expression was inflammatory cytokines-inducible, and most of the studies focused on inflammation diseases. Increasing number of cancer studies began to reveal its biological role in cancers recently, although with contradictory findings in literature. It was discovered from our earlier prostate cancer cell line models studies that when prostate cancer cells treated with either ethidium bromide or a cell cycle inhibitor flavopiridol for a long-term, the treatment-survived tumor cells experienced metabolic reprogramming toward Warburg effect pathways with greater aggressive features, and one common finding from these cells was the upregulation of GBP1. In this study, possible role of GBP1 in two independent prostate cancer lines by application of CRISR/Cas9 gene knockout (KO) technology was investigated. The GBP1 gene KO DU145 and PC3 prostate cancer cells were significantly less aggressive in vitro, with less proliferation, migration, wound healing, and colony formation capabilities, in addition to a significantly lower level of mitochondrial oxidative phosphorylation and glycolysis. At the same time, such GBP1 KO cells were significantly more sensitive to chemotherapeutic reagents. Xenograft experiments verified a significantly slower tumor growth of the GBP1 KO cells in nude mouse model. Furthermore, GBP1 protein expression in clinical prostate cancer sample revealed its aggressive clinical feature correlation and shorter overall survival association. Collectively, our results indicate a pro-survival or oncogenic role of GBP1 in prostate cancer.

5.
Onco Targets Ther ; 11: 3149-3157, 2018.
Article in English | MEDLINE | ID: mdl-29881288

ABSTRACT

BACKGROUND: Sushi repeat-containing protein X-linked 2 (SRPX2) is overexpressed in a variety of different tumor tissues and correlated with poor prognosis in patients. Little research focuses on the role of SRPX2 expression in prostate cancer (PCa), and the clinicopathological significance of the protein expression in this tumor is relatively unknown. However, our previous transcriptome data from those cancer stem-like cells indicated the role of SRPX2 in PCa. MATERIALS AND METHODS: In this study, RT-PCR and Western blotting were firstly used to examine the SRPX2 expression in three PCa cell lines including LNCaP, DU145, and PC3, and then SRPX2 protein expression was immunohistochemically investigated and statistically analyzed in a series of 106 paraffin-embedded PCa tissue specimens. RESULTS: Significantly lower levels of SRPX2 expression were verified in the LNCaP cells, compared with the expression in the aggressive DU145 and PC3 cells, in both mRNA and protein levels. Immunohistochemically, there were variable SRPX2 protein expressions in the clinical samples. Moreover, high levels of SRPX2 expression in the PCa tissues were significantly associated with Gleason score (P=0.008), lymph node metastasis (P=0.009), and distant metastasis (P=0.021). Furthermore, higher levels of SRPX2 expression in the PCa tissues were significantly associated with shorter overall survival (OS) (P<0.001). CONCLUSION: Our results demonstrate that SRPX2 is highly expressed in aggressive PCa cells in vitro, and its protein expression in PCa is significantly associated with malignant clinical features and shorter OS, strongly indicating its prognostic value in prostate cancers.

6.
RSC Adv ; 8(30): 16636-16644, 2018 May 03.
Article in English | MEDLINE | ID: mdl-35540547

ABSTRACT

Although abnormal metabolism in metabolic syndrome and tumours has been well described, the relationship between oxoglutarate dehydrogenase (OGDH) and obesity-related diseases is still largely unknown. This study aimed to investigate whether it was possible to use transcription activator-like effector nuclease (TALEN) technology to establish OGDH-/- rats and then study the effect of a high-fat diet (HFD) on these rats. However, after OGDH+/-rats were generated, we were unable to identify any OGDH-/- rats by performing mating experiments with the OGDH+/- rats for almost one year. During the past three years, only OGDH+/- rats were stably established, and correspondingly reduced OGDH expression in the tissues of the OGDH+/- rats was verified. No significant abnormal behaviour was observed in the OGDH+/- rats compared to the wild-type (WT) control rats. However, the OGDH+/- rats were revealed to have higher body weight, and the difference was even significantly greater under the HFD condition. Furthermore, blood biochemical and tissue histological examinations uncovered no abnormalities with normal diets, but a HFD resulted in liver dysfunction with pathological alterations in the OGDH+/- rats. Our results strongly indicate that OGDH homologous knockout is lethal in rats but heterologous OGDH knockout results in vulnerable liver lesions with a HFD. Therefore, the current study may provide a useful OGDH+/- rat model for further investigations of metabolic syndrome and obesity-related hepatic carcinogenesis.

7.
Tumour Biol ; 39(8): 1010428317713671, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28789597

ABSTRACT

Our earlier study revealed that long-term ethidium bromide application causes mitochondrial DNA depletion in human prostate cancer DU145 cell line (DU145MtDP), and this DU145MtDP subline appears to have expanded CD44Bright cell population than its parental wild type DU145 cells (DU145WT). Increasing evidence suggests that CD44Bright cells are highly cancer stem cell like, but it is not clear about their dynamic transition between CD44Dim and CD44Bright phenotypes in prostate cancer cells, and how it is affected by mitochondrial DNA depletion. To address these questions, four cell subpopulations were isolated from both DU145WT and DU145MtDP cell lines based on their CD44 expression level and mitochondrial membrane potential. The cell motility and colony formation capability of the fluorescence activated cell sorting-sorted cell subpopulations were further examined. It was discovered in the DU145WT cells that CD44Dim cells could transit into both CD44Dim and CD44Bright phenotypes and that CD44Bright cells were prone to sustain their CD44Bright phenotype as renewal. However, such transition principle was altered in the DU145MtDP cells, in which CD44Bright cells showed similar capability to sustain a CD44Bright phenotype, while the transition of CD44Dim cells to CD44Bright were suppressed. It is concluded that mitochondrial DNA depletion in the human prostate cancer DU145 cells influences their renewal and CD44 subphenotype transition. Such alterations may be the driving force for the enrichment of CD44Bright DU145 cells after the mitochondrial DNA depletion, although the molecular mechanisms remain unclear.


Subject(s)
Cell Lineage/genetics , Cell Proliferation/genetics , DNA, Mitochondrial/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Ethidium/pharmacology , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Humans , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Male , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/pathology
8.
Sci Rep ; 7(1): 5081, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698547

ABSTRACT

Flavopiridol (FP) is a pan-cyclin dependent kinase inhibitor, which shows strong efficacy in inducing cancer cell apoptosis. Although FP is potent against most cancer cells in vitro, unfortunately it proved less efficacious in clinical trials in various aggressive cancers. To date, the molecular mechanisms of the FP resistance are mostly unknown. Here, we report that a small fraction human prostate cancer DU145 cells can survive long-term FP treatment and emerge as FP-resistant cells (DU145FP). These DU145FP cells show accumulated mitochondrial lesions with stronger glycolytic features, and they proliferate in slow-cycling and behave highly migratory with strong anti-apoptotic potential. In addition, the cells are less sensitive to cisplatin and docetaxel-induced apoptotic pressure, and over-express multiple stem cell associated biomarkers. Our studies collectively uncover for the first time that FP-resistant prostate cancer cells show metabolic remodeling, and the metabolic plasticity might be required for the FP resistance-associated cancer cell stemness up-regulation.


Subject(s)
Drug Resistance, Neoplasm , Flavonoids/therapeutic use , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/pharmacology , Docetaxel/pharmacology , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Piperidines/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Pseudopodia/drug effects , Pseudopodia/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Oncotarget ; 8(28): 46363-46380, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28624784

ABSTRACT

One of the remarkable features of cancer cells is aerobic glycolysis, a phenomenon known as the "Warburg Effect", in which cells rely preferentially on glycolysis instead of oxidative phosphorylation (OXPHOS) as the main energy source even in the presence of high oxygen tension. Cells with dysfunctional mitochondria are unable to generate sufficient ATP from mitochondrial OXPHOS, and then are forced to rely on glycolysis for ATP generation. Here we report our results in a prostate cancer cell line in which the mitochondrial pyruvate carrier 1 (MPC1) gene was knockout. It was discovered that the MPC1 gene knockout cells revealed a metabolism reprogramming to aerobic glycolysis with reduced ATP production, and the cells became more migratory and resistant to both chemotherapy and radiotherapy. In addition, the MPC1 knockout cells expressed significantly higher levels of the stemness markers Nanog, Hif1α, Notch1, CD44 and ALDH. To further verify the correlation of MPC gene function and cell stemness/metabolic reprogramming, MPC inhibitor UK5099 was applied in two ovarian cancer cell lines and similar results were obtained. Taken together, our results reveal that functional MPC may determine the fate of metabolic program and the stemness status of cancer cells in vitro.


Subject(s)
Energy Metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Animals , Anion Transport Proteins , Biomarkers , Cell Line, Tumor , Citric Acid Cycle , DNA Mutational Analysis , Disease Models, Animal , Gene Expression Regulation , Glucose/metabolism , Glycolysis , Humans , Male , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters , Mutation , Neoplasms/genetics , Oxidative Phosphorylation , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
11.
Oncotarget ; 8(8): 13344-13356, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28076853

ABSTRACT

Cells generate adenosine-5'-triphosphate (ATP), the major currency for energy-consuming reactions, through mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis. One of the remarkable features of cancer cells is aerobic glycolysis, also known as the "Warburg Effect", in which cancer cells rely preferentially on glycolysis instead of mitochondrial OXPHOS as the main energy source even in the presence of high oxygen tension. One of the main players in controlling OXPHOS is the mitochondrial gatekeeperpyruvate dehydrogenase complex (PDHc) and its major subunit is E1α (PDHA1). To further analyze the function of PDHA1 in cancer cells, it was knock out (KO) in the human prostate cancer cell line LnCap and a stable KO cell line was established. We demonstrated that PDHA1 gene KO significantly decreased mitochondrial OXPHOS and promoted anaerobic glycolysis, accompanied with higher stemness phenotype including resistance to chemotherapy, enhanced migration ability and increased expression of cancer stem cell markers. We also examined PDHA1 protein expression in prostate cancer tissues by immunohistochemistry and observed that reduced PDHA1 protein expression in clinical prostate carcinomas was significantly correlated with poor prognosis. Collectively, our results show that negative PDHA1 gene expressionis associated with significantly higher cell stemness in prostate cancer cells and reduced protein expression of this gene is associated with shorter clinical outcome in prostate cancers.


Subject(s)
Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Aged , Aged, 80 and over , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Gene Knockout Techniques , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplastic Stem Cells/enzymology , Polymerase Chain Reaction , Prognosis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/mortality
12.
Oncotarget ; 8(1): 1058-1073, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27911865

ABSTRACT

Aerobic glycolysis is one of the emerging hallmarks of cancer cells. In this study, we investigated the relationship between blocking mitochondrial pyruvate carrier (MPC) with MPC blocker UK5099 and the metabolic alteration as well as aggressive features of esophageal squamous carcinoma. It was found that blocking pyruvate transportation into mitochondria attenuated mitochondrial oxidative phosphorylation (OXPHOS) and triggered aerobic glycolysis, a feature of Warburg effect. In addition, the HIF-1α expression and ROS production were also activated upon UK5099 application. It was further revealed that the UK5099-treated cells became significantly more resistant to chemotherapy and radiotherapy, and the UK5099-treated tumor cells also exhibited stronger invasive capacity compared to the parental cells. In contrast to esophageal squamous epithelium cells, decreased MPC protein expression was observed in a series of 157 human squamous cell carcinomas, and low/negative MPC1 expression predicted an unfavorable clinical outcome. All these results together revealed the potential connection of altered MPC expression/activity with the Warburg metabolic reprogramming and tumor aggressiveness in cell lines and clinical samples. Collectively, our findings highlighted a therapeutic strategy targeting Warburg reprogramming of human esophageal squamous cell carcinomas.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Phenotype , Adenosine Triphosphate/metabolism , Biological Transport , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma , Glucose/metabolism , Glycolysis/drug effects , Humans , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters , Neoplasm Metastasis , Neoplasm Staging , Oxidation-Reduction , Prognosis , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
13.
BMC Cancer ; 16(1): 894, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27852261

ABSTRACT

BACKGROUND: Cancer cells exhibit an altered metabolism, which is characterized by a preference for aerobic glycolysis more than mitochondrial oxidation of pyruvate. Mitochondrial pyruvate carrier 1 (MPC1) and mitochondrial pyruvate carrier 2 (MPC2) play a bottleneck role by transporting pyruvate into mitochondrial through the mitochondrial inner membrane. Therefore, their protein expression in cancers may be of clinical consequences. There are studies showing low levels of MPC1 expression in colon, kidney and lung cancers, and the expression of MPC1 correlates with poor prognosis. However, the expression status of MPC1 and MPC2 in prostate cancer (PCA) is unclear. METHODS: In this study, expression of MPC1 and MPC2 in LNCaP and DU145 prostate cancer cell lines was examined by immunocytochemistry (ICC) and Western blotting. Compared to the LNCaP cells, lower levels of MPC1 and MPC2 expression in the DU145 cell line was identified. We then extended our study to 88 patients with prostate cancer who underwent transurethral electro-vaporization of prostate or radical prostatectomy at the First Affiliated Hospital of Zhengzhou University, Henan, China. Patient-derived paraffin embedded PCA specimens were collected for immunohistochemistry (IHC). Correlations with clinicopathologic factors were evaluated by Chi-square or Fisher´s exact probability tests. Overall survival (OS) rates were determined using the Kaplan-Meier estimator. The Cox proportional hazard regression model was used in univariate analysis and multivariate analysis to identify factors significantly correlated with prognosis. RESULTS: Linear regression analysis revealed that MPC1 expression level was positively correlated with MPC2 expression (r = 0.375, P = 0.006) in the prostate cancers. MPC1 expression was negatively associated with UICC stage (P = 0.031). While UICC stage (P < 0.001) and lymph node metastasis (P = 0.002) were negatively associated with MPC2 expression. Positive MPC1 or MPC2 expression in cancer tissues was significantly associated with higher OS (P < 0.05). The multivariate analysis showed that both MPC1 and MPC2 expressions in PCA were independent prognostic factors for higher OS (For MPC1: RR = 0.654, 95% CI: 0.621-0690, P < 0.001; For MPC2: RR = 0.696, 95% CI: 0.660-0.734, P < 0.001). CONCLUSIONS: Our study indicates that MPC1 and MPC2 expressions are of prognostic values in PCAs and that positive expression of MPC1 or MPC2 is a predictor of favorable outcome.


Subject(s)
Anion Transport Proteins/genetics , Gene Expression , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Aged , Aged, 80 and over , Cell Line , Follow-Up Studies , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Mitochondria/metabolism , Monocarboxylic Acid Transporters , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Prostatic Neoplasms/diagnosis
14.
Am J Cancer Res ; 6(9): 2076-2087, 2016.
Article in English | MEDLINE | ID: mdl-27725912

ABSTRACT

Pyruvate dehydrogenase A1 (PDHA1) serves as a gate-keeper enzyme link between glycolysis and the mitochondrial citric acid cycle. The inhibition of PDHA1 in cancer cells can result in an increased Warburg effect and a more aggressive phenotype in cancer cells. This study was conducted to investigate the expression of PDHA1 in ovarian cancer and the correlation between PDHA1 expression and the prognosis of patients. The PDHA1 protein expression in 3 ovarian cancer cell lines (OVCAR-3, SKOV-3 and ES-2) and 248 surgically removed ovarian carcinoma samples was immunocytochemically examined. Statistical analyses were performed to evaluate the correlations between PDHA1 expression and the clinicopathological characteristics of the patients as well as the predictive value of PDHA1. The results showed the presence of variable expression of PDHA1 in the three ovarian cancer cell lines. Of the 248 ovarian cancer tissue specimens, 45 cases (18.1%) were negative in tumor cells for PDHA1, 162 cases (65.3%) displayed a low expression level, and 41 cases (16.5%) had a relatively high PDHA1 staining. The expression of PDHA1 was associated with the histological subtype (P=0.004) and FIGO stage (P=0.002). The median OS time in the PDHA1 negative group, low expression group and high expression group were 0.939 years, 1.443 years and 9.900 years, respectively. The median PFS time in the above three groups were 0.287 years, 0.586 years and 9.900 years, respectively. Furthermore, the high expression of PDHA1 in ovarian carcinoma cells was significantly associated with better OS and PFS by statistical analyses. Multivariate analyses showed that PDHA1 expression was also an independent prognostic factor for higher OS in ovarian cancer patients (HR=0.705, 95% CI 0.541-0.918, P=0.01). Our study indicated that the decreased expression of PDHA1 might be an independent prognostic factor in unfavorable outcomes.

15.
Am J Cancer Res ; 6(9): 2088-2097, 2016.
Article in English | MEDLINE | ID: mdl-27725913

ABSTRACT

Ovarian cancer is the most lethal gynecologic malignancy, in which cancer stem cells (CSC) have been reported to be the driving force of relapse and therapy-resistance. It is therefore important to explore CSC markers in ovarian cancer. This project aimed to explore the correlation between the expression of potential CSC maker Cacna2d1 and clinicopathological parameters in 238 epithelial ovarian cancer (EOC) samples. Immunohistochemically, positive Cacna2d1 expression was observed in 83.6% (199/238) of the EOC tumors, among which 107 tumors (44.9%) were highly positive and 92 (38.7%) tumors were weakly positive for the Cacna2d1 protein expression. Among the 158 serous carcinomas, the Cacna2d1 positivity was 148 (93.7%), in which 88 (55.7%) were highly positive, and 60 (38.0%) were weakly positive for the Cacna2d1 protein expression. Most strikingly, the Cacna2d1 was specifically expressed in the infiltration front areas of the EOC tumors. Statistical analyses showed that positive expression of Cacna2d1 was significantly associated with advanced FIGO stage (P<0.001), histological subtype (P=0.017) and tumor differentiation (P=0.015). Positive Cacna2d1 protein expression was significantly associated with poor overall survival (OS) and shorter progression free survival (PFS) in both total EOCs and serous carcinomas, although multivariate analyses did not reach statistical significance. In summary, our results suggest Cacna2d1 protein may play a crucial role in promoting aggressive EOC behavior and progression, and Cacna2d1 may serve as a novel predictive prognostic marker and a potential target for therapeutic intervention in EOCs.

16.
Oncotarget ; 7(33): 53837-53852, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27462778

ABSTRACT

Alternative pathways of metabolism endowed cancer cells with metabolic stress. Inhibiting the related compensatory pathways might achieve synergistic anticancer results. This study demonstrated that pyruvate dehydrogenase E1α gene knockout (PDHA1 KO) resulted in alterations in tumor cell metabolism by rendering the cells with increased expression of glutaminase1 (GLS1) and glutamate dehydrogenase1 (GLUD1), leading to an increase in glutamine-dependent cell survival. Deprivation of glutamine induced cell growth inhibition, increased reactive oxygen species and decreased ATP production. Pharmacological blockade of the glutaminolysis pathway resulted in massive tumor cells apoptosis and dysfunction of ROS scavenge in the LNCaP PDHA1 KO cells. Further examination of the key glutaminolysis enzymes in human prostate cancer samples also revealed that higher levels of GLS1 and GLUD1 expression were significantly associated with aggressive clinicopathological features and poor clinical outcome. These insights supply evidence that glutaminolysis plays a compensatory role for cell survival upon alternative energy metabolism and targeting the glutamine anaplerosis of energy metabolism via GLS1 and GLUD1 in cancer cells may offer a potential novel therapeutic strategy.


Subject(s)
Glutamate Dehydrogenase/metabolism , Glutaminase/metabolism , Glutamine/metabolism , Prostatic Neoplasms/metabolism , Aged , Gene Knockout Techniques , Humans , Male , Middle Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Pyruvate Dehydrogenase (Lipoamide)/metabolism
17.
Oncotarget ; 7(26): 40297-40313, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27248169

ABSTRACT

Reducing mtDNA content was considered as a critical step in the metabolism restructuring for cell stemness restoration and further neoplastic development. However, the connections between mtDNA depletion and metabolism reprograming-based cancer cell stemness in prostate cancers are still lack of studies. Here, we demonstrated that human CRPC cell line PC3 tolerated high concentration of the mtDNA replication inhibitor ethidium bromide (EtBr) and the mtDNA depletion triggered a universal metabolic remodeling process. Failure in completing that process caused lethal consequences. The mtDNA depleted (MtDP) PC3 cells could be steadily maintained in the special medium in slow cycling status. The MtDP PC3 cells contained immature mitochondria and exhibited Warburg effect. Furthermore, the MtDP PC3 cells were resistant to therapeutic treatments and contained greater cancer stem cell-like subpopulations: CD44+, ABCG2+, side-population and ALDHbright. In conclusion, these results highlight the association of mtDNA content, mitochondrial function and cancer cell stemness features.


Subject(s)
DNA, Mitochondrial/genetics , Neoplastic Stem Cells/cytology , Prostatic Neoplasms/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Cell Count , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , DNA Replication/drug effects , Ethidium/chemistry , Humans , Hyaluronan Receptors/metabolism , Hypoxia/metabolism , Male , Neoplasm Proteins/metabolism , Oxygen/chemistry , Oxygen Consumption , Prostatic Neoplasms/genetics
18.
World J Surg Oncol ; 14: 63, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26940557

ABSTRACT

BACKGROUND: Abdominoperineal excision is performed in patients with locally advanced, low rectal carcinoma. Reconstruction of the dorsal vagina and perineum using the vertical rectus abdominis myocutaneous flap following extensive surgery results in favorable surgical outcome and quality of life. However, the rectus abdominis muscle, as part of the anterior abdominal wall, may develop fibrous lesions also as a transplant. CASE PRESENTATION: A 39-year-old female patient with low rectal cancer and extensive colorectal polyposis was treated with neoadjuvant chemoradiotherapy followed by colectomy and abdominoperineal excision with resection of the dorsal vaginal wall and subsequent reconstruction of the perineum using the vertical rectus abdominis myocutaneous flap. At the 6-month follow-up, a suspected 2 × 2 cm tumor recurrence was detected in the transposed tissue and was subsequently surgically removed. Histologic examination concluded with fibromatosis. Genetic testing revealed a known disease-causing mutation in the adenomatous polyposis coli gene, confirming the diagnosis of familial adenomatous polyposis. CONCLUSIONS: Fibromatosis may affect the anterior abdominal wall, that is the rectus abdominis muscle, at the primary site or may develop in the muscle after its transposition into the perineum at pelvic reconstruction. Fibromatosis in the muscle flap after pelvic reconstruction may present a difficult diagnostic challenge for the multidisciplinary team.


Subject(s)
Fibroma/diagnosis , Myocutaneous Flap/pathology , Neoplasm Recurrence, Local/diagnosis , Rectal Neoplasms/surgery , Rectus Abdominis/pathology , Adult , Diagnosis, Differential , Female , Fibroma/etiology , Humans , Neoplasm Recurrence, Local/etiology , Prognosis , Rectal Neoplasms/complications
19.
Anticancer Res ; 36(1): 343-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26722063

ABSTRACT

BACKGROUND: Aldehyde dehydrogenase-1 (ALDH1) has been shown to be a potential cancer stem cell marker in different types of cancer. However, the role of its expression in tumor cells and the microenvironment in different types of cancer is still controversial. MATERIALS AND METHODS: ALDH1 protein was immunohistochemically investigated and analyzed in 157 samples of surgically dissected esophageal squamous cell carcinoma (ESCC) tissues. RESULTS: ALDH1 protein expression in ESCC tumor cells was significantly associated with poor differentiation (p<0.05) and strongly positive ALDH1 expression in tumor cells was related with shorter overall survival (p<0.05), while the expression of ALDH1 in ESCC stromal cells had no significant relationship with clinicopathological features (p>0.05). CONCLUSION: High expression of cancer stem cell marker ALDH1 in ESCC cells may thus portend a poor prognosis. However, its expression in the tumor microenvironment did not appear to have a role in ESCC behavior. More studies are warranted to find out the mechanisms to explain this.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/therapy , Isoenzymes/metabolism , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Treatment Outcome
20.
APMIS ; 123(12): 1040-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26547370

ABSTRACT

Zinc finger E-box-binding homeobox 1 (ZEB1) is a transcriptional factor known to repress E-cadherin promoter and thus induce EMT. Expression of ZEB-1 has in numerous cancers been associated with aggressive disease and poor clinical outcome. Our aim was to investigate the expression of ZEB1 in esophageal squamous- and small-cell carcinomas. Immunohistochemical staining was performed on tissue sections obtained from 151 patients with esophageal squamous cell carcinoma (ESCC) and 25 patients with primary small-cell carcinoma of the esophagus (PSCCE). Semi-quantitative analysis, and thus statistical analysis, has been accomplished on the samples. Immunohistochemistry revealed ZEB1 expression in the cytoplasm (64.9% of cases), in nuclei (11.3% of cases) and in tumor stroma (80.1% of cases) of ESCC. In PSCCE only nuclear staining (88.0% of cases) was observed. Weak cytoplasmic expression of ZEB1 in ESCC was associated with longer survival. Immunohistochemical evaluation of ZEB1 cytoplasmic expression in ESCC may have clinical prognostic value, but further studies are needed to fully understand the function as well as potential clinical and therapeutic implications of ZEB1 expression in cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Small Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Esophageal Neoplasms/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Esophageal Squamous Cell Carcinoma , Esophagus/pathology , Humans , Immunohistochemistry , Middle Aged , Staining and Labeling , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL