Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Commun ; 13(1): 6168, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36257965

ABSTRACT

Actively dividing cells, including some cancers, rely on aerobic glycolysis rather than oxidative phosphorylation to generate energy, a phenomenon termed the Warburg effect. Constitutive activation of the Hypoxia Inducible Factor (HIF-1), a transcription factor known for mediating an adaptive response to oxygen deprivation (hypoxia), is a hallmark of the Warburg effect. HIF-1 is thought to promote glycolysis and suppress oxidative phosphorylation. Here, we instead show that HIF-1 can promote gluconeogenesis. Using a multiomics approach, we reveal the genomic, transcriptomic, and metabolomic landscapes regulated by constitutively active HIF-1 in C. elegans. We use RNA-seq and ChIP-seq under aerobic conditions to analyze mutants lacking EGL-9, a key negative regulator of HIF-1. We integrate these approaches to identify over two hundred genes directly and functionally upregulated by HIF-1, including the PEP carboxykinase PCK-1, a rate-limiting mediator of gluconeogenesis. This activation of PCK-1 by HIF-1 promotes survival in response to both oxidative and hypoxic stress. Our work identifies functional direct targets of HIF-1 in vivo, comprehensively describing the metabolome induced by HIF-1 activation in an organism.


Subject(s)
Caenorhabditis elegans , Gluconeogenesis , Animals , Caenorhabditis elegans/genetics , Gluconeogenesis/genetics , Transcription Factors/genetics , Cell Hypoxia , Hypoxia/genetics , Oxygen , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
3.
Cell Rep ; 17(9): 2445-2459, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27840052

ABSTRACT

Extensive transcriptional and ontogenetic diversity exists among normal tissue-resident macrophages, with unique transcriptional profiles endowing the cells with tissue-specific functions. However, it is unknown whether the origins of different macrophage populations affect their roles in malignancy. Given potential artifacts associated with irradiation-based lineage tracing, it remains unclear if bone-marrow-derived macrophages (BMDMs) are present in tumors of the brain, a tissue with no homeostatic involvement of BMDMs. Here, we employed multiple models of murine brain malignancy and genetic lineage tracing to demonstrate that BMDMs are abundant in primary and metastatic brain tumors. Our data indicate that distinct transcriptional networks in brain-resident microglia and recruited BMDMs are associated with tumor-mediated education yet are also influenced by chromatin landscapes established before tumor initiation. Furthermore, we demonstrate that microglia specifically repress Itga4 (CD49D), enabling its utility as a discriminatory marker between microglia and BMDMs in primary and metastatic disease in mouse and human.


Subject(s)
Brain Neoplasms/pathology , Macrophages/pathology , Animals , Base Sequence , Bone Marrow Cells/pathology , Brain Neoplasms/genetics , Cell Lineage , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioma/genetics , Glioma/pathology , Humans , Integrin alpha4/metabolism , Macrophage Activation , Macrophages/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Sequence Analysis, RNA , Transcription Factors/metabolism
4.
Nat Med ; 19(10): 1264-72, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24056773

ABSTRACT

Glioblastoma multiforme (GBM) comprises several molecular subtypes, including proneural GBM. Most therapeutic approaches targeting glioma cells have failed. An alternative strategy is to target cells in the glioma microenvironment, such as tumor-associated macrophages and microglia (TAMs). Macrophages depend on colony stimulating factor-1 (CSF-1) for differentiation and survival. We used an inhibitor of the CSF-1 receptor (CSF-1R) to target TAMs in a mouse proneural GBM model, which significantly increased survival and regressed established tumors. CSF-1R blockade additionally slowed intracranial growth of patient-derived glioma xenografts. Surprisingly, TAMs were not depleted in treated mice. Instead, glioma-secreted factors, including granulocyte-macrophage CSF (GM-CSF) and interferon-γ (IFN-γ), facilitated TAM survival in the context of CSF-1R inhibition. Expression of alternatively activated M2 markers decreased in surviving TAMs, which is consistent with impaired tumor-promoting functions. These gene signatures were associated with enhanced survival in patients with proneural GBM. Our results identify TAMs as a promising therapeutic target for proneural gliomas and establish the translational potential of CSF-1R inhibition for GBM.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Macrophages/cytology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Brain Neoplasms/metabolism , Disease Progression , Glioblastoma/metabolism , Mice , Signal Transduction
5.
Cancer Res ; 71(19): 6174-83, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21844184

ABSTRACT

Although the molecular changes that characterize gliomas have been studied, the pathogenesis of tumor development remains unclear. p21 contributes to gliomagenesis by stabilizing cyclin D1-cdk4 kinase complexes, suggesting that cyclin D1 and cdk4 may also be required for glial tumor development. In this study, we used a mouse model to attempt to confirm this hypothesis, finding that cyclin D1 and cdk4 played active roles in not only the tumor but also the tumor microenvironment. Loss of cdk4 blocked tumor development, but loss of cyclin D1 did not prevent gliomas from developing. Instead, loss of cyclin D1 impeded progression to higher stages of malignancy. Enforcing expression of cyclin D1 was insufficient to correct the progression defect observed in cyclin D1-deficient animals. In contrast, restoration of cdk4 in the cdk4-deficient animals restored cell proliferation and tumor formation, although at lower tumor grades. Notably, the failure of tumors in the cyclin D1- and cdk4-deficient animals to progress to higher grades was correlated with a failure to fully activate microglia in the tumor microenvironment. Moreover, when platelet-derived growth factor-transformed glial cells were engrafted orthotopically into the mice, the tumors that formed progressed to high grades in wild-type mice but not cyclin D1-deficient animals. Together, our findings establish that the cyclin D1-cdk4 axis is not only critical in glial tumor cells but also in stromal-derived cells in the surrounding tumor microenvironment that are vital to sustain tumor outgrowth.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Oligodendroglioma/etiology , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Chickens , Mice , Mice, Knockout , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Tumor Microenvironment
6.
J Neurosci ; 29(33): 10299-308, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19692604

ABSTRACT

The adult mammalian brain responds to injury by activating a program of cell proliferation during which many oligodendrocyte precursors, microglia, and some astrocytes proliferate. Another common response to brain injury is the induction of reactive gliosis, a process whereby dormant astrocytes undergo morphological changes and alter their transcriptional profiles. Although brain injury-induced reactive gliosis is concurrent with the proliferation of surrounding cells, a functional relationship between reactive gliosis and this cell proliferation has not been clearly demonstrated. Here, we show that the mitogen sonic hedgehog (SHH) is produced in reactive astrocytes after injury to the cerebral cortex and participates in regulating the proliferation of Olig2-expressing (Olig2(+)) cells after brain injury. Using a cortical freeze injury to induce reactive gliosis in a Gli-luciferase reporter mouse, we show that the SHH pathway is maximally active 3 d after brain injury and returns to baseline levels by 14 d. SHH expression parallels Gli activation and localizes to glial fibrillary acidic protein-expressing reactive astrocytes. Inhibition of the SHH pathway with cyclopamine blocks the Gli response and significantly reduces both the proliferating and overall number of Olig2(+) cells in the injured cortex. To provide mechanistic insight into SHH pathway activation in astrocytes, we show that proinflammatory stimuli activate SHH-expressing reactive astrocytes, whereas inhibition of inflammation-induced reactive gliosis by macrophage depletion abolishes SHH activation after brain injury and dampens cell proliferation after injury. Our data describes a unique reactive astrocyte-based, SHH-expressing niche formed in response to injury and inflammation that regulates the proliferation of Olig2(+) cells.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Hedgehog Proteins/physiology , Inflammation Mediators/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/metabolism , Inflammation Mediators/antagonists & inhibitors , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/biosynthesis , Oligodendrocyte Transcription Factor 2
SELECTION OF CITATIONS
SEARCH DETAIL
...