Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Cancer Lett ; 559: 216104, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36863507

ABSTRACT

Hepatoid adenocarcinoma (HAC) is a rare, malignant, extrahepatic tumor with histologic features similar to those of hepatocellular carcinoma. HAC is most often associated with elevated alpha-fetoprotein (AFP). HAC can occur in multiple organs, including the stomach, esophagus, colon, pancreas, lungs, and ovaries. HAC differs greatly from typical adenocarcinoma in terms of its biological aggression, poor prognosis, and clinicopathological characteristics. However, the mechanisms underlying its development and invasive metastasis remain unclear. The purpose of this review was to summarize the clinicopathological features, molecular traits, and molecular mechanisms driving the malignant phenotype of HAC, in order to support the clinical diagnosis and treatment of HAC.


Subject(s)
Adenocarcinoma , Carcinoma, Hepatocellular , Liver Neoplasms , Stomach Neoplasms , Humans , Liver Neoplasms/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/drug therapy , Carcinoma, Hepatocellular/pathology , alpha-Fetoproteins
2.
J Biol Chem ; 296: 100369, 2021.
Article in English | MEDLINE | ID: mdl-33545174

ABSTRACT

Previous studies have identified GAPDH as a promising target for treating cancer and modulating immunity because its inhibition reduces glycolysis in cells (cancer cells and immune cells) with the Warburg effect, a modified form of cellular metabolism found in cancer cells. However, the quantitative relationship between GAPDH and the aerobic glycolysis remains unknown. Here, using siRNA-mediated knockdown of GAPDH expression and iodoacetate-dependent inhibition of enzyme activity, we examined the quantitative relationship between GAPDH activity and glycolysis rate. We found that glycolytic rates were unaffected by the reduction of GAPDH activity down to 19% ± 4.8% relative to untreated controls. However, further reduction of GAPDH activity below this level caused proportional reductions in the glycolysis rate. GAPDH knockdown or inhibition also simultaneously increased the concentration of glyceraldehyde 3-phosphate (GA3P, the substrate of GAPDH). This increased GA3P concentration countered the effect of GAPDH knockdown or inhibition and stabilized the glycolysis rate by promoting GAPDH activity. Mechanistically, the intracellular GA3P concentration is controlled by the Gibbs free energy of the reactions upstream of GAPDH. The thermodynamic state of the reactions along the glycolysis pathway was only affected when GAPDH activity was reduced below 19% ± 4.8%. Doing so moved the reactions catalyzed by GAPDH + PGK1 (phosphoglycerate kinase 1, the enzyme immediate downstream of GAPDH) away from the near-equilibrium state, revealing an important biochemical basis to interpret the rate control of glycolysis by GAPDH. Collectively, we resolved the numerical relationship between GAPDH and glycolysis in cancer cells with the Warburg effect and interpreted the underlying mechanism.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/physiology , Glycolysis/physiology , Neoplasms/metabolism , Cell Line, Tumor , Glucose/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Oxidation-Reduction , RNA, Small Interfering/genetics , Warburg Effect, Oncologic
3.
Sci Rep ; 4: 4927, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24820099

ABSTRACT

Warburg effect is a dominant phenotype of most cancer cells. Here we show that this phenotype depends on its environment. When cancer cells are under regular culture condition, they show Warburg effect; whereas under lactic acidosis, they show a nonglycolytic phenotype, characterized by a high ratio of oxygen consumption rate over glycolytic rate, negligible lactate production and efficient incorporation of glucose carbon(s) into cellular mass. These two metabolic modes are intimately interrelated, for Warburg effect generates lactic acidosis that promotes a transition to a nonglycolytic mode. This dual metabolic nature confers growth advantage to cancer cells adapting to ever changing microenvironment.


Subject(s)
Acidosis, Lactic/metabolism , Neoplasms/metabolism , Animals , Biological Transport , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Glycolysis , Heterografts , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Mice , NAD/metabolism , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Oxygen Consumption , Phenotype
4.
PLoS One ; 8(1): e52706, 2013.
Article in English | MEDLINE | ID: mdl-23300986

ABSTRACT

PURPOSE: Cancer drug resistance is a major obstacle for the success of chemotherapy. Since most clinical anticancer drugs could induce drug resistance, it is desired to develop candidate drugs that are highly efficacious but incompetent to induce drug resistance. Numerous previous studies have proven that shikonin and its analogs not only are highly tumoricidal but also can bypass drug-transporter and apoptotic defect mediated drug resistance. The purpose of this study is to investigate if or not shikonin is a weak inducer of cancer drug resistance. EXPERIMENTAL DESIGN: Different cell lines (K562, MCF-7, and a MDR cell line K562/Adr), after repeatedly treated with shikonin for 18 months, were assayed for drug resistance and gene expression profiling. RESULTS: After 18-month treatment, cells only developed a mere 2-fold resistance to shikonin and a marginal resistance to cisplatin and paclitaxel, without cross resistance to shikonin analogs and other anticancer agents. Gene expression profiles demonstrated that cancer cells did strongly respond to shikonin treatment but failed to effectively mobilize drug resistant machineries. Shikonin-induced weak resistance was associated with the up-regulation of ßII-tubulin, which physically interacted with shikonin. CONCLUSION: Taken together, apart from potent anticancer activity, shikonin and its analogs are weak inducers of cancer drug resistance and can circumvent cancer drug resistance. These merits make shikonin and its analogs potential candidates for cancer therapy with advantages of avoiding induction of drug resistance and bypassing existing drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Naphthoquinones/pharmacology , Neoplasms/drug therapy , Cell Proliferation , Cisplatin/pharmacology , Drug Resistance, Multiple/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , MCF-7 Cells , Neoplasms/pathology , Paclitaxel/pharmacology , Time Factors
5.
World J Gastrointest Oncol ; 4(6): 145-51, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22737276

ABSTRACT

AIM: To explore the value of serum M2-pyruvate kinase (M2-PK) in colorectal cancer (CRC) mass screening. METHODS: We conducted a molecular epidemiology study in Hangzhou, China, from year 2006 to year 2008. Serum samples were collected from 93 CRC, 41 advanced adenomas, 137 adenomas, 47 non-adenomatous polyps, and 158 normal participants in a community setting. Serum M2-PK and carcinoembryonic antigen (CEA) were measured using Enzyme-linked immunosorbent assay. SPSS 16.0 software was used to perform data analysis. Area under the receiver operating characteristic curve (AUC), sensitivity, and specificities were estimated for serum M2-PK in diagnosis of colorectal lesions and compared with CEA. RESULTS: Average serum M2-PK value among 158 normal people was 2.96 U/mL and not affected by gender (P = 0.47) or age (P = 0.59). Average serum M2-PK (U/mL) was 14.75 among stage III and 13.10 among stage I and II CRC patients, about 4 times higher than that among normal people. Average serum M2-PK was 8.58, 6.70, 5.13 and 2.51 U/mL among advanced adenoma, adenomas, non-adenomatous polyps, and inflammatory bowel disease patients, respectively. AUC for serum M2-PK was greater than that for CEA among all colorectal lesions. AUC for serum M2-PK was 0.89 (0.84, 0.94) (95% confidence interval), higher than that for CEA [0.70 (0.62-0.79)] in CRC stage I and II, 0.89 (0.84-0.94) vs 0.73 (0.63-0.83) in CRC stage III, 0.81 (0.74-0.86) vs 0.63 (0.53 - 0.73) in advanced adenomas, 0.69 (0.64-0.76) vs 0.54 (0.47-0.60) in adenomas, and 0.69 (0.62-0.78) vs 0.58 (0.48-0.68) in non-adenomatous polyps. The diagnostic sensitivity for all colorectal lesions increased with decrease in the cut-off value of serum M2-PK. The diagnostic sensitivity (%) of serum M2-PK was 100.00 for CRC, 95.12 advanced adenoma, 82.48 adenoma, and 82.98 non-adenomatous polyp. There were no CRC cases missed and 40.51% of unnecessary colonoscopies were avoided when the cut-off value was 2.00 U/mL. CONCLUSION: Serum M2-PK can be used as a primary screening test in CRC mass screening. It may be a promising non-invasive biomarker for CRC early detection.

6.
PLoS One ; 6(12): e28335, 2011.
Article in English | MEDLINE | ID: mdl-22164272

ABSTRACT

BACKGROUND: To mitigate the cardiotoxicity of anthracycline antibiotics without compromising their anticancer activities is still an issue to be solved. We previously demonstrated that schisandrin B (Sch B) could protect against doxorubicin (Dox)-induced acute cardiotoxicity via enhancing cardiomyocytic glutathione redox cycling that could attenuate oxidative stress generated from Dox. In this study, we attempted to prove if Sch B could also protect against Dox-induced chronic cardiotoxicity, a more clinically relevant issue, without compromising its anticancer activity. METHODOLOGY: Rat was given intragastrically either vehicle or Sch B (50 mg/kg) two hours prior to i.p. Dox (2.5 mg/kg) weekly over a 5-week period with a cumulative dose of Dox 12.5 mg/kg. At the 6th and 12th week after last dosing, rats were subjected to cardiac function measurement, and left ventricles were processed for histological and ultrastructural examination. Dox anticancer activity enhanced by Sch B was evaluated by growth inhibition of 4T1, a breast cancer cell line, and S180, a sarcoma cell line, in vitro and in vivo. PRINCIPAL FINDINGS: Pretreatment with Sch B significantly attenuated Dox-induced loss of cardiac function and damage of cardiomyocytic structure. Sch B substantially enhanced Dox cytotoxicities toward S180 in vitro and in vivo in mice, and increased Dox cytotoxcity against 4T1 in vitro. Although we did not observe this enhancement against the implanted 4T1 primary tumor, the spontaneous metastasis to lung was significantly reduced in combined treatment group than Dox alone group. CONCLUSION: Sch B is capable of protecting Dox-induced chronic cardiotoxicity and enhancing its anticancer activity. To the best of our knowledge, Sch B is the only molecule ever proved to function as a cardioprotective agent as well as a chemotherapeutic sensitizer, which is potentially applicable for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/toxicity , Lignans/pharmacology , Polycyclic Compounds/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation , Cyclooctanes/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Glutathione/metabolism , Heart/drug effects , Heart Ventricles/drug effects , Male , Mice , Mice, Inbred BALB C , Myocardium/pathology , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Sprague-Dawley
7.
Clin Cancer Res ; 13(22 Pt 1): 6753-60, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006777

ABSTRACT

PURPOSE: The dose-cumulative cardiotoxicities and the emerging cancerous apoptotic/drug resistance are two major obstacles limiting the efficacy of anthracycline antibiotics, notably doxorubicin. We attempted to prove if schisandrin B (Sch B), a dual inhibitor of P-glycoprotein and multidrug resistance-associated protein 1, could protect against doxorubicin-induced cardiotoxicity, on the premise that Sch B is an enhancer of glutathione redox cycling that may attenuate doxorubicin-induced oxidative stress in the cardiomyocytes. EXPERIMENTAL DESIGN: Mice or rat were dosed with a single injection of doxorubicin (25 mg/kg, i.p.) with or without pretreatment of Sch B. The protective roles of Sch B against doxorubicin-induced cardiac damage were evaluated on the aspects of the release of cardiac enzymes into serum, the formation of malondialdehyde, the activation of matrix metalloproteinase, the structural damage in the left ventricles, the mortality rates, and the cardiac functions. RESULTS: Pretreatment of Sch B significantly attenuated doxorubicin-induced cardiotoxicities on all the aspects listed above. The underlying mechanism was associated with the effect of Sch B on maintaining the cardiomyocytic glutathione and the activities of superoxide dismutase, and the key enzymes (glutathione peroxidase, glutathione reductase, and glutathione transferase) responsible for glutathione redox cycling, which neutralized doxorubicin-induced oxidative stress. CONCLUSION: To the best of our knowledge, Sch B is the only molecule ever proved to function as a cardioprotective agent as well as a dual inhibitor of P-glycoprotein and multidrug resistance-associated protein 1, which is potentially applicable to treat cancers, especially the multidrug-resistant cancers involving doxorubicin or its kin.


Subject(s)
Anthracyclines/antagonists & inhibitors , Antibiotics, Antineoplastic/antagonists & inhibitors , Doxorubicin/antagonists & inhibitors , Glutathione/metabolism , Heart Diseases/prevention & control , Lignans/therapeutic use , Polycyclic Compounds/therapeutic use , Animals , Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Cyclooctanes/therapeutic use , Doxorubicin/toxicity , Heart Diseases/chemically induced , Mice , Mice, Inbred ICR , Oxidation-Reduction
8.
Mol Cancer Ther ; 6(5): 1641-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17513612

ABSTRACT

Defect in apoptotic signaling and up-regulation of drug transporters in cancer cells significantly limits the effectiveness of cancer chemotherapy. We propose that an agent inducing non-apoptotic cell death may overcome cancer drug resistance and showed that shikonin, a naturally occurring naphthoquinone, induced a cell death in MCF-7 and HEK293 distinct from apoptosis and characterized with (a) a morphology of necrotic cell death; (b) loss of plasma membrane integrity; (c) loss of mitochondrial membrane potentials; (d) activation of autophagy as a downstream consequence of cell death, but not a contributing factor; (e) elevation of reactive oxygen species with no critical roles contributing to cell death; and (f) that the cell death was prevented by a small molecule, necrostatin-1, that specifically prevents cells from necroptosis. The characteristics fully comply with those of necroptosis, a basic cell-death pathway recently identified by Degterev et al. with potential relevance to human pathology. Furthermore, we proved that shikonin showed a similar potency toward drug-sensitive cancer cell lines (MCF-7 and HEK293) and their drug-resistant lines overexpressing P-glycoprotein, Bcl-2, or Bcl-x(L), which account for most of the clinical cancer drug resistance. To our best knowledge, this is the first report to document the induction of necroptosis by a small molecular compound to circumvent cancer drug resistance.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Naphthoquinones/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Death , Dose-Response Relationship, Drug , Female , HeLa Cells , Humans , Mice , Mice, Nude , Necrosis , Neoplasm Transplantation
9.
Syst Appl Microbiol ; 30(3): 207-12, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16908113

ABSTRACT

A bacterial strain HLK1(T) was isolated from the human erythroleukemia cell line K562. This bacterium is a Gram-negative rod, motile with a polar flagellum. It is strictly aerobic, nonfermentative, and oxidase and catalase positive. Its optimal growth occurs at 37 degrees C at pH between 6.5 and 7.5. Phylogenetically, although it shares 98% similarity with the 16S rRNA of Phenylobacterium lituiforme, the DNA-DNA hybridization value between the two species is only 43%. HLK1(T) has a DNA G+C content of 71.2+/-0.2 mol%. It is a facultative intracellular organism and may have pathogenic relevance with humans and mammals. On the basis of the phylogenetic and phenotypic characterization, strain HLK1(T) is proposed to be classified in the genus Phenylobacterium, as P. zucineum sp. nov. The type strain is HLK1(T) (=CGMCC 1.3786(T), DSM=18354).


Subject(s)
Caulobacteraceae/classification , Caulobacteraceae/isolation & purification , K562 Cells/microbiology , Caulobacteraceae/genetics , DNA Gyrase/genetics , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , HeLa Cells , Humans , Molecular Sequence Data , Phylogeny , RNA, Ribosomal, 16S/genetics
10.
Life Sci ; 80(8): 741-8, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17129588

ABSTRACT

We recently reported that dibenzocyclooctadiene lignans were a novel class of P-glycoprotein (P-gp) inhibitors. In this study, we demonstrated that the lignans of this class were also effective inhibitors of multidrug resistance-associated protein 1 (MRP1). The activities of 5 dibenzocyclooctadiene lignans (schisandrin A, schisandrin B, schisantherin A, schisandrol A, and schisandrol B) to reverse MRP1-mediated drug resistance were tested using HL60/Adriamycin (ADR) and HL60/Multidrug resistance-associated protein (MRP), two human promyelocytic leukemia cell lines with overexpression of MRP1 but not P-gp. The five lignans could effectively reverse drug resistance of the two cell lines to vincristine, daunorubicin, and VP-16. This study, together with our previous reports, proves that dibenzocyclooctadiene lignans have multiple activities against cancer multidrug resistance, including inhibition of P-gp and MRP1, and enhancement of apoptosis. Considering that cancer multidrug resistance (MDR) is multifactorial, agents with broad activities are preferable to the use of combination of several specific modulators to prevent drug-drug interaction and cumulative toxicity.


Subject(s)
Cyclooctanes/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm/drug effects , Lignans/pharmacology , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclooctanes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HL-60 Cells/drug effects , HL-60 Cells/metabolism , Humans , Lignans/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL