Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Cancer Drug Targets ; 17(2): 158-168, 2017.
Article in English | MEDLINE | ID: mdl-27262320

ABSTRACT

The evolution of genomic research enabled the genetic and molecular profiling of breast cancer and revealed the profound complexity and heterogeneity of this disease. Subtypes of breast cancer characterized by mutations and/or amplifications of some proto-oncogenes are associated with an increased rate of recurrence and poor prognosis. They represent a challenge in the clinic with limited arsenal to attack them. Nowadays, metabolic reprogramming is firmly established as a hallmark of cancer. An increased rate of lipid and protein syntheses in cancerous tissues, a direct consequence of alterations in key metabolic enzymes involved in these pathways, is now recognized as an important aspect of the rewired metabolism of neoplastic cells. Over the past several years, accumulating evidence has revealed that mutations or amplifications of some proto-oncogenes are primarily involved in this metabolic dysregulation. It is thus critically important to dissect the molecular mechanisms tumors use to link metabolic reprogramming with upstream altered signaling. In this article, we review the recent findings that support the importance of lipid and protein biosyntheses in breast tumorigenesis, discuss the crosstalk between growth factor signal transduction and key metabolic enzymes involved in these processes, and point out the potentials of developing new strategies and therapeutics to target these key parameters in order to help breast cancer patients by providing new therapeutic opportunities.


Subject(s)
Breast Neoplasms/metabolism , Enzymes/metabolism , Lipid Metabolism , Proteins/metabolism , Breast Neoplasms/drug therapy , Female , Humans , Molecular Targeted Therapy/methods , Protein Biosynthesis , Receptor, ErbB-2/metabolism
2.
Antimicrob Agents Chemother ; 60(9): 5285-93, 2016 09.
Article in English | MEDLINE | ID: mdl-27324775

ABSTRACT

Isoniazid (INH) remains the core drug in tuberculosis management, but serious hepatotoxicity and potentially fatal liver injury continue to accompany INH consumption. Among numerous theories that have been established to explain INH-induced liver injury, an inflammatory stress theory has recently been widely used to explain the idiosyncrasy. Inflammatory stress usually sensitizes tissues to a drug's toxic consequences. Therefore, the present study was conducted to verify whether bacterial lipopolysaccharide (LPS)-induced inflammation may have a role in enhancing INH hepatotoxicity. While single INH or LPS administration showed no major toxicity signs, INH-LPS cotreatment intensified liver toxicity. Both blood biomarkers and histological evaluations clearly showed positive signs of severe liver damage accompanied by massive necrosis, inflammatory infiltration, and hepatic steatosis. Furthermore, elevated serum levels of bile acid associated with the repression of bile acid synthesis and transport regulatory parameters were observed. Moreover, the principal impact of cytochrome P450 2E1 (CYP2E1) on INH toxicity could be anticipated, as its protein expression showed enormous increases in INH-LPS-cotreated animals. Furthermore, the crucial role of CYP2E1 in the production of reactive oxygen species (ROS) was clearly obvious in the repression of hepatic antioxidant parameters. In summary, these results confirmed that this LPS-induced inflammation model might prove valuable in revealing the hepatotoxic mechanisms of INH and the crucial role played by CYP2E1 in the initiation and propagation of INH-induced liver damage, information which could be very useful to clinicians in understanding the pathogenesis of drug-induced liver injury.


Subject(s)
Antitubercular Agents/adverse effects , Chemical and Drug Induced Liver Injury/enzymology , Cytochrome P-450 CYP2E1/genetics , Fatty Liver/enzymology , Isoniazid/adverse effects , Lipopolysaccharides/toxicity , Animals , Bile Acids and Salts/blood , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Drug Combinations , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression , Inflammation , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/pathology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Up-Regulation
3.
Oncotarget ; 6(26): 21865-77, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26068969

ABSTRACT

Increased lipogenesis and protein synthesis is a hallmark of cancer cell proliferation, survival, and metastatic progression and is under intense investigation as a potential antineoplastic target. Acetyltanshinone IIA (ATA) is a compound that was obtained from chemical modifications of tanshinone IIA (TIIA), a potent anticancer agent extracted from the dried roots of the Chinese herbal medicine Salvia miltiorrhiza Bunge. A previous investigation indicated that ATA is more effective in inhibiting the growth of breast cancer especially cells with HER2 overexpression. However, the molecular mechanism(s) mediating this cytotoxic effect on HER2-positive breast cancer remained undefined. Studies described here report that ATA induced G1/S phase arrest and apoptosis in the HER2-positive MDA-MB-453, SK-BR-3, and BT-474 breast cancer cell lines. Mechanistic investigations revealed that the ATA-induced apoptosis effect is associated with remarkably down-regulation of receptor tyrosine kinases (RTKs) EGFR/HER2 and inhibition of their downstream pro-survival signaling pathways. Interestingly, ATA was found to trigger oxidative and endoplasmic reticulum (ER) stresses and to activate AMP activated protein kinase (AMPK) leading to inactivation of key enzymes involved in lipid and protein biogenesis. Intraperitoneal administration of ATA significantly inhibited the growth of MDA-MB-453 xenografts in athymic mice without causing weight loss and any other side effects. Additionally, transwell migration, invasion, and wound healing assays revealed that ATA could suppress tumor angiogenesis in vitro. Taken together, our data suggest that ATA may have broad utility in the treatment of HER2-overexpressed breast cancers.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Phenanthrenes/pharmacology , Receptor, ErbB-2/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Down-Regulation , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Humans , Lipids/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress/drug effects , Protein Biosynthesis/drug effects , Random Allocation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...