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.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30794774

ABSTRACT

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Subject(s)
Intestinal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/metabolism , Cell Line , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Deoxycholic Acid/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Intestinal Neoplasms/genetics , Intestines , Liver , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/physiology , Organoids/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Risk Factors , Signal Transduction , Taurocholic Acid/analogs & derivatives , Taurocholic Acid/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
2.
Cell Rep ; 22(10): 2521-2529, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29514081

ABSTRACT

PGC1α is a pleiotropic co-factor that affects angiogenesis, mitochondrial biogenesis, and oxidative muscle remodeling via its association with multiple transcription factors, including the master oxidative nuclear receptor ERRγ. To decipher their epistatic relationship, we explored ERRγ gain of function in muscle-specific PGC1α/ß double-knockout (PKO) mice. ERRγ-driven transcriptional reprogramming largely rescues muscle damage and improves muscle function in PKO mice, inducing mitochondrial biogenesis, antioxidant defense, angiogenesis, and a glycolytic-to-oxidative fiber-type transformation independent of PGC1α/ß. Furthermore, in combination with voluntary exercise, ERRγ gain of function largely restores mitochondrial energetic deficits in PKO muscle, resulting in a 5-fold increase in running performance. Thus, while PGC1s can interact with multiple transcription factors, these findings implicate ERRs as the major molecular target through which PGC1α/ß regulates both innate and adaptive energy metabolism.


Subject(s)
Mitochondria/metabolism , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Nuclear Proteins/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Animals , Energy Metabolism , Mice, Knockout , Oxidation-Reduction
3.
Cell Metab ; 25(5): 1186-1193.e4, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28467934

ABSTRACT

Management of energy stores is critical during endurance exercise; a shift in substrate utilization from glucose toward fat is a hallmark of trained muscle. Here we show that this key metabolic adaptation is both dependent on muscle PPARδ and stimulated by PPARδ ligand. Furthermore, we find that muscle PPARδ expression positively correlates with endurance performance in BXD mouse reference populations. In addition to stimulating fatty acid metabolism in sedentary mice, PPARδ activation potently suppresses glucose catabolism and does so without affecting either muscle fiber type or mitochondrial content. By preserving systemic glucose levels, PPARδ acts to delay the onset of hypoglycemia and extends running time by ∼100 min in treated mice. Collectively, these results identify a bifurcated PPARδ program that underlies glucose sparing and highlight the potential of PPARδ-targeted exercise mimetics in the treatment of metabolic disease, dystrophies, and, unavoidably, the enhancement of athletic performance.


Subject(s)
Glucose/metabolism , PPAR delta/metabolism , Physical Endurance , Running , Animals , Cell Line , Fatty Acids/metabolism , Male , Mice , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Physical Conditioning, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...