Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Sci Adv ; 7(15)2021 04.
Article de Anglais | MEDLINE | ID: mdl-33827825

RÉSUMÉ

The serine/threonine kinase ULK1 mediates autophagy initiation in response to various cellular stresses, and genetic deletion of ULK1 leads to accumulation of damaged mitochondria. Here we identify Parkin, the core ubiquitin ligase in mitophagy, and PARK2 gene product mutated in familial Parkinson's disease, as a ULK1 substrate. Recent studies uncovered a nine residue ("ACT") domain important for Parkin activation, and we demonstrate that AMPK-dependent ULK1 rapidly phosphorylates conserved serine108 in the ACT domain in response to mitochondrial stress. Phosphorylation of Parkin Ser108 occurs maximally within five minutes of mitochondrial damage, unlike activation of PINK1 and TBK1, which is observed thirty to sixty minutes later. Mutation of the ULK1 phosphorylation sites in Parkin, genetic AMPK or ULK1 depletion, or pharmacologic ULK1 inhibition, all lead to delays in Parkin activation and defects in assays of Parkin function and downstream mitophagy events. These findings reveal an unexpected first step in the mitophagy cascade.

2.
Nat Med ; 22(10): 1108-1119, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27643638

RÉSUMÉ

Continuous de novo fatty acid synthesis is a common feature of cancer that is required to meet the biosynthetic demands of a growing tumor. This process is controlled by the rate-limiting enzyme acetyl-CoA carboxylase (ACC), an attractive but traditionally intractable drug target. Here we provide genetic and pharmacological evidence that in preclinical models ACC is required to maintain the de novo fatty acid synthesis needed for growth and viability of non-small-cell lung cancer (NSCLC) cells. We describe the ability of ND-646-an allosteric inhibitor of the ACC enzymes ACC1 and ACC2 that prevents ACC subunit dimerization-to suppress fatty acid synthesis in vitro and in vivo. Chronic ND-646 treatment of xenograft and genetically engineered mouse models of NSCLC inhibited tumor growth. When administered as a single agent or in combination with the standard-of-care drug carboplatin, ND-646 markedly suppressed lung tumor growth in the Kras;Trp53-/- (also known as KRAS p53) and Kras;Stk11-/- (also known as KRAS Lkb1) mouse models of NSCLC. These findings demonstrate that ACC mediates a metabolic liability of NSCLC and that ACC inhibition by ND-646 is detrimental to NSCLC growth, supporting further examination of the use of ACC inhibitors in oncology.


Sujet(s)
Acetyl-coA carboxylase/antagonistes et inhibiteurs , Carcinome pulmonaire non à petites cellules/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Acides gras/biosynthèse , Métabolisme lipidique/effets des médicaments et des substances chimiques , Tumeurs du poumon/métabolisme , Pyrimidinones/pharmacologie , Thiophènes/pharmacologie , AMP-Activated Protein Kinases , Acetyltransferases/antagonistes et inhibiteurs , Régulation allostérique , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Prolifération cellulaire/génétique , Humains , Métabolisme lipidique/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Souris , Souris knockout , Thérapie moléculaire ciblée , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes p21(ras)/génétique , Protéine p53 suppresseur de tumeur/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Mol Cancer Ther ; 10(3): 427-36, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-21282354

RÉSUMÉ

Many types of cancer cells require a supply of fatty acids (FA) for growth and survival, and interrupting de novo FA synthesis in model systems causes potent anticancer effects. We hypothesized that, in addition to synthesis, cancer cells may obtain preformed, diet-derived FA by uptake from the bloodstream. This would require hydrolytic release of FA from triglyceride in circulating lipoprotein particles by the secreted enzyme lipoprotein lipase (LPL), and the expression of CD36, the channel for cellular FA uptake. We find that selected breast cancer and sarcoma cells express and secrete active LPL, and all express CD36. We further show that LPL, in the presence of triglyceride-rich lipoproteins, accelerates the growth of these cells. Providing LPL to prostate cancer cells, which express low levels of the enzyme, did not augment growth, but did prevent the cytotoxic effect of FA synthesis inhibition. Moreover, LPL knockdown inhibited HeLa cell growth. In contrast to the cell lines, immunohistochemical analysis confirmed the presence of LPL and CD36 in the majority of breast, liposarcoma, and prostate tumor tissues examined (n = 181). These findings suggest that, in addition to de novo lipogenesis, cancer cells can use LPL and CD36 to acquire FA from the circulation by lipolysis, and this can fuel their growth. Interfering with dietary fat intake, lipolysis, and/or FA uptake will be necessary to target the requirement of cancer cells for FA.


Sujet(s)
Prolifération cellulaire , Matières grasses alimentaires/métabolisme , Acides gras/métabolisme , Lipoprotein lipase/métabolisme , Tumeurs/métabolisme , Animaux , Antigènes CD36/génétique , Lignée cellulaire tumorale , Inhibiteurs de la synthèse d'acides gras/pharmacologie , Acides gras/pharmacologie , Femelle , Humains , Lipolyse , Liposarcome/génétique , Liposarcome/métabolisme , Mâle , Souris , Souris transgéniques , Séquençage par oligonucléotides en batterie , Petit ARN interférent/génétique
4.
Int J Oncol ; 36(5): 1309-14, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20372807

RÉSUMÉ

Liposarcomas (LS) are mesenchymal tumors that can recur after surgical resection and often do not respond to presently available medical therapies. This study demonstrates the dependence of LS on de novo long-chain fatty acid synthesis for growth. Lipogenesis can be impaired by inhibiting the activities of lipogenic enzymes, including acetyl CoA-carboxylase (ACC) and fatty acid synthase (FASN), or by suppressing the expression of key genes involved in the pathway and its regulation. The FASN inhibitors cerulenin and orlistat reduced the growth of two LS cell lines (LiSa2, SW872), as did inhibition of ACC with soraphen A. CDDO-Me, a synthetic triterpenoid, suppressed expression of Spot 14 and FASN genes and likewise inhibited LS cell growth. Importantly, the anti-proliferative effect of each agent was prevented by the co-administration of palmitate, the major product of cellular long-chain fatty acid synthesis. In stark contrast to LS cells, these compounds had no effect on the growth of fibroblasts. Four biochemically distinct agents that target critical points in the fatty acid synthetic pathway exert anti-proliferative effects on LS cells, and rescue of cell growth by palmitic acid suggests that reduced tumor cell lipogenesis mediates the growth inhibition. These findings warrant further studies aimed at the clinical exploitation of the dependence of LS cell growth on fatty acids.


Sujet(s)
Acides gras/composition chimique , Régulation de l'expression des gènes tumoraux , Liposarcome/métabolisme , Acetyl-coA carboxylase/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Relation dose-effet des médicaments , Fatty acid synthases/métabolisme , Fibroblastes/métabolisme , Humains , Lactones/pharmacologie , Lipides/composition chimique , Orlistat , RT-PCR
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE