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.
Nat Commun ; 11(1): 4055, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792504

ABSTRACT

Although metastasis is the most common cause of cancer deaths, metastasis-intrinsic dependencies remain largely uncharacterized. We previously reported that metastatic pancreatic cancers were dependent on the glucose-metabolizing enzyme phosphogluconate dehydrogenase (PGD). Surprisingly, PGD catalysis was constitutively elevated without activating mutations, suggesting a non-genetic basis for enhanced activity. Here we report a metabolic adaptation that stably activates PGD to reprogram metastatic chromatin. High PGD catalysis prevents transcriptional up-regulation of thioredoxin-interacting protein (TXNIP), a gene that negatively regulates glucose import. This allows glucose consumption rates to rise in support of PGD, while simultaneously facilitating epigenetic reprogramming through a glucose-fueled histone hyperacetylation pathway. Restoring TXNIP normalizes glucose consumption, lowers PGD catalysis, reverses hyperacetylation, represses malignant transcripts, and impairs metastatic tumorigenesis. We propose that PGD-driven suppression of TXNIP allows pancreatic cancers to avidly consume glucose. This renders PGD constitutively activated and enables metaboloepigenetic selection of additional traits that increase fitness along glucose-replete metastatic routes.


Subject(s)
Chromatin/metabolism , Glucose/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Biological Transport/genetics , Biological Transport/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Chromatin Immunoprecipitation , Epigenesis, Genetic/genetics , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Phosphogluconate Dehydrogenase/genetics , Phosphogluconate Dehydrogenase/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
2.
Oncogene ; 37(38): 5248-5256, 2018 09.
Article in English | MEDLINE | ID: mdl-29849117

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) adopts several unique metabolic strategies to support primary tumor growth. Whether additional metabolic strategies are adopted to support metastatic tumorigenesis is less clear. This could be particularly relevant for distant metastasis, which often follows a rapidly progressive clinical course. Here we report that PDAC distant metastases evolve a unique series of metabolic reactions to maintain activation of the anabolic glucose enzyme phosphogluconate dehydrogenase (PGD). PGD catalytic activity was recurrently elevated across distant metastases, and modulating PGD activity levels dictated tumorigenic capacity. Metabolomics data raised the possibility that distant metastases evolved a core pentose conversion pathway (PCP) that converted glucose-derived metabolites into PGD substrate, thereby hyperactivating the enzyme. Consistent with this, each individual metabolite in the PCP stimulated PGD catalysis in distant metastases, and knockdown of each individual PCP enzyme selectively impaired tumorigenesis. We propose that the PCP manufactures PGD substrate outside of the rate-limiting oxidative pentose phosphate pathway (oxPPP). This enables PGD-dependent tumorigenesis by providing adequate substrate to fuel high catalytic activity, and raises the possibility that PDAC distant metastases adopt their own unique metabolic strategies to support tumor growth.


Subject(s)
Carcinogenesis , Pancreatic Neoplasms/pathology , Pentose Phosphate Pathway , Cell Line, Tumor , Humans , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms
3.
Nat Genet ; 49(3): 367-376, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28092686

ABSTRACT

During the progression of pancreatic ductal adenocarcinoma (PDAC), heterogeneous subclonal populations emerge that drive primary tumor growth, regional spread, distant metastasis, and patient death. However, the genetics of metastases largely reflects that of the primary tumor in untreated patients, and PDAC driver mutations are shared by all subclones. This raises the possibility that an epigenetic process might operate during metastasis. Here we report large-scale reprogramming of chromatin modifications during the natural evolution of distant metastasis. Changes were targeted to thousands of large chromatin domains across the genome that collectively specified malignant traits, including euchromatin and large organized chromatin histone H3 lysine 9 (H3K9)-modified (LOCK) heterochromatin. Remarkably, distant metastases co-evolved a dependence on the oxidative branch of the pentose phosphate pathway (oxPPP), and oxPPP inhibition selectively reversed reprogrammed chromatin, malignant gene expression programs, and tumorigenesis. These findings suggest a model whereby linked metabolic-epigenetic programs are selected for enhanced tumorigenic fitness during the evolution of distant metastasis.


Subject(s)
Epigenesis, Genetic/genetics , Glucose/metabolism , Neoplasm Metastasis/genetics , Pancreatic Neoplasms/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Chromatin/genetics , Epigenomics/methods , Gene Expression/genetics , Heterochromatin/genetics , Histones/genetics , Humans , Pancreatic Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...