Hedgehog-YAP Signaling Pathway Regulates Glutaminolysis to Control Activation of Hepatic Stellate Cells

Autor: Steve S. Choi, Jeongeun Hyun, Marzena Swiderska-Syn, Richard T. Premont, Youngmi Jung, Anna Mae Diehl, Bahar Salimian Rizi, George D. Dalton, Kuo Du, Gregory A. Michelotti, Eric Thelen
Rok vydání: 2017
Předmět:
0301 basic medicine
Liver Cirrhosis
Time Factors
Glutamine
Cell Cycle Proteins
Mitochondria
Liver

Liver Cirrhosis
Experimental

chemistry.chemical_compound
Myofibroblasts
Cells
Cultured

YAP1
Mice
Knockout

Chemistry
Transdifferentiation
Gastroenterology
Cellular Reprogramming
Smoothened Receptor
Hedgehog signaling pathway
Phenotype
Editorial
Liver
Ketoglutaric Acids
RNA Interference
Signal Transduction
Cyclopamine
Transfection
03 medical and health sciences
Glutaminase
Hepatic Stellate Cells
Animals
Humans
Hedgehog Proteins
Hedgehog
Adaptor Proteins
Signal Transducing

Cell Proliferation
Glutaminolysis
Hepatology
YAP-Signaling Proteins
Phosphoproteins
Rats
Mice
Inbred C57BL

030104 developmental biology
Gene Expression Regulation
Case-Control Studies
Cell Transdifferentiation
Cancer research
Hepatic stellate cell
Smoothened
Energy Metabolism
Transcription Factors
Zdroj: Cellular and Molecular Gastroenterology and Hepatology
ISSN: 1528-0012
Popis: Background & Aims Cirrhosis results from accumulation of myofibroblasts derived from quiescent hepatic stellate cells (Q-HSCs); it regresses when myofibroblastic HSCs are depleted. Hedgehog signaling promotes transdifferentiation of HSCs by activating Yes-associated protein 1 (YAP1 or YAP) and inducing aerobic glycolysis. However, increased aerobic glycolysis alone cannot meet the high metabolic demands of myofibroblastic HSCs. Determining the metabolic processes of these cells could lead to strategies to prevent progressive liver fibrosis, so we investigated whether glutaminolysis (conversion of glutamine to alpha-ketoglutarate) sustains energy metabolism and permits anabolism when Q-HSCs become myofibroblastic, and whether this is controlled by hedgehog signaling to YAP. Methods Primary HSCs were isolated from C57BL/6 or Smo flox/flox mice; we also performed studies with rat and human myofibroblastic HSCs. We measured changes of glutaminolytic genes during culture-induced primary HSC transdifferentiation. Glutaminolysis was disrupted in cells by glutamine deprivation or pathway inhibitors (bis-2-[5-phenylacetamido-1,2,4-thiadiazol-2-yl] ethyl sulfide, CB-839, epigallocatechin gallate, and aminooxyacetic acid), and effects on mitochondrial respiration, cell growth and migration, and fibrogenesis were measured. Hedgehog signaling to YAP was disrupted in cells by adenovirus expression of Cre-recombinase or by small hairpin RNA knockdown of YAP. Hedgehog and YAP activity were inhibited by incubation of cells with cyclopamine or verteporfin, and effects on glutaminolysis were measured. Acute and chronic liver fibrosis were induced in mice by intraperitoneal injection of CCl 4 or methionine choline-deficient diet. Some mice were then given injections of bis-2-[5-phenylacetamido-1,2,4-thiadiazol-2-yl] ethyl sulfide to inhibit glutaminolysis, and myofibroblast accumulation was measured. We also performed messenger RNA and immunohistochemical analyses of percutaneous liver biopsies from healthy human and 4 patients with no fibrosis, 6 patients with mild fibrosis, and 3 patients with severe fibrosis. Results Expression of genes that regulate glutaminolysis increased during transdifferentiation of primary Q-HSCs into myofibroblastic HSCs, and inhibition of glutaminolysis disrupted transdifferentiation. Blocking glutaminolysis in myofibroblastic HSCs suppressed mitochondrial respiration, cell growth and migration, and fibrogenesis; replenishing glutaminolysis metabolites to these cells restored these activities. Knockout of the hedgehog signaling intermediate smoothened or knockdown of YAP inhibited expression of glutaminase, the rate-limiting enzyme in glutaminolysis. Hedgehog and YAP inhibitors blocked glutaminolysis and suppressed myofibroblastic activities in HSCs. In livers of patients and of mice with acute or chronic fibrosis, glutaminolysis was induced in myofibroblastic HSCs. In mice with liver fibrosis, inhibition of glutaminase blocked accumulation of myofibroblasts and fibrosis progression. Conclusions Glutaminolysis controls accumulation of myofibroblast HSCs in mice and might be a therapeutic target for cirrhosis.
Databáze: OpenAIRE