Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1860(11 Pt A): 2363-2376, 2016 11.
Article in English | MEDLINE | ID: mdl-27418237

ABSTRACT

BACKGROUND: Recent studies have shown that low density lipoproteins reconstituted with the natural omega 3 fatty acid docosahexaenoic acid (LDL-DHA) is selectively cytotoxic to liver cancer cells over normal hepatocytes. To date, little is known about the subcellular events which transpire following LDL-DHA treatment. METHODS: Herein, murine noncancer and cancer liver cells, TIB-73 and TIB-75 respectively, were investigated utilizing confocal microscopy, flow cytometry and viability assays to demonstrate differential actions of LDL-DHA nanoparticles in normal versus malignant cells. RESULTS: Our studies first showed that basal levels of oxidative stress are significantly higher in the malignant TIB-75 cells compared to the normal TIB-73 cells. As such, upon entry of LDL-DHA into the malignant TIB-75 cells, DHA is rapidly oxidized precipitating global and lysosomal lipid peroxidation along with increased lysosomal permeability. This leakage of lysosomal contents and lipid peroxidation products trigger subsequent mitochondrial dysfunction and nuclear injury. The cascade of LDL-DHA mediated lipid peroxidation and organelle damage was partially reversed by the administration of the antioxidant, N-acetylcysteine, or the iron-chelator, deferoxamine. LDL-DHA treatment in the normal TIB-73 cells was well tolerated and did not elicit any cell or organelle injury. CONCLUSION: These studies have shown that LDL-DHA is selectively cytotoxic to liver cancer cells and that increased levels of ROS and iron catalyzed reactions promote the peroxidation of DHA which lead to organelle dysfunction and ultimately the demise of the cancer cell. GENERAL SIGNIFICANCE: LDL-DHA selectively disrupts lysosomal, mitochondrial and nuclear function in cancer cells as a novel pathway for eliminating cancer cells.


Subject(s)
Docosahexaenoic Acids/pharmacology , Hepatocytes/metabolism , Nanoparticles , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Line , Cell Line, Tumor , Cells, Cultured , DNA Damage , Docosahexaenoic Acids/toxicity , Hepatocytes/drug effects , Humans , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/toxicity , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress
2.
Gastroenterology ; 150(2): 488-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26484708

ABSTRACT

BACKGROUND & AIMS: Dietary intake of the natural omega-3 fatty acid docosahexaenoic acid (DHA) has been implicated in protecting patients with viral hepatitis B or C from developing hepatocellular carcinoma (HCC). Little is known about the effects of DHA on established solid tumors. Here we describe a low-density lipoprotein-based nanoparticle that acts as a transporter for unesterified DHA (LDL-DHA) and demonstrates selective cytotoxicity toward HCC cells. We investigated the ability of LDL-DHA to reduce growth of orthotopic hepatomas in rats. METHODS: AxC-Irish (ACI) rats were given intrahepatic injections of rat hepatoma cells (H4IIE); 24 tumor-bearing rats (mean tumor diameter, ∼1 cm) were subject to a single hepatic artery injection of LDL nanoparticles (2 mg/kg) loaded with DHA (LDL-DHA), triolein (LDL-TO), or sham surgery controls. Tumor growth was measured by magnetic resonance imaging and other methods; tumor, liver, and serum samples were collected and assessed by histochemical, immunofluorescence, biochemical, and immunoblot analyses. RESULTS: Three days after administration of LDL-TO or sham surgery, the control rats had large, highly vascularized tumors that contained proliferating cells. However, rats given LDL-DHA had smaller, pale tumors that were devoid of vascular supply and >80% of the tumor tissue was necrotic. Four to 6 days after injection of LDL-DHA, the tumors were 3-fold smaller than those of control rats. The liver tissue that surrounded the tumors showed no histologic or biochemical evidence of injury. Injection of LDL-DHA into the hepatic artery of rats selectively deregulated redox reactions in tumor tissues by increasing levels of reactive oxygen species and lipid peroxidation, depleting and oxidizing glutathione and nicotinamide adenine dinucleotide phosphate, and significantly down-regulating the antioxidant enzyme glutathione peroxidase-4. Remarkably, the redox balance in the surrounding liver was not disrupted. CONCLUSION: LDL-DHA nanoparticle selectively kills hepatoma cells and reduces growth of orthotopic liver tumors in rats. It induces tumor-specific necrosis by selectively disrupting redox balance within the cancer cell.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Docosahexaenoic Acids/administration & dosage , Drug Carriers , Lipoproteins, LDL/administration & dosage , Liver Neoplasms/drug therapy , Nanoparticles , Animals , Antineoplastic Agents/metabolism , Antioxidants/metabolism , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Docosahexaenoic Acids/metabolism , Dose-Response Relationship, Drug , Hepatic Artery , Infusions, Intra-Arterial , Lipid Peroxidation/drug effects , Lipoproteins, LDL/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Necrosis , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Time Factors , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...