Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 46
1.
bioRxiv ; 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38617314

How genetic lesions drive cell transformation and whether they can be circumvented without compromising function of non-transformed cells are enduring questions in oncology. Here we show that in mature T cells-in which physiologic clonal proliferation is a cardinal feature- constitutive MYC transcription and Tsc1 loss in mice modeled aggressive human malignancy by reinforcing each other's oncogenic programs. This cooperation was supported by MYC-induced large neutral amino acid transporter chaperone SLC3A2 and dietary leucine, which in synergy with Tsc1 deletion overstimulated mTORC1 to promote mitochondrial fitness and MYC protein overexpression in a positive feedback circuit. A low leucine diet was therapeutic even in late-stage disease but did not hinder T cell immunity to infectious challenge, nor impede T cell transformation driven by constitutive nutrient mTORC1 signaling via Depdc5 loss. Thus, mTORC1 signaling hypersensitivity to leucine as an onco-nutrient enables an onco-circuit, decoupling pathologic from physiologic utilization of nutrient acquisition pathways.

2.
Cell Stem Cell ; 31(2): 161-180, 2024 02 01.
Article En | MEDLINE | ID: mdl-38306993

Stem cells perform many different functions, each of which requires specific metabolic adaptations. Over the past decades, studies of pluripotent and tissue stem cells have uncovered a range of metabolic preferences and strategies that correlate with or exert control over specific cell states. This review aims to describe the common themes that emerge from the study of stem cell metabolism: (1) metabolic pathways supporting stem cell proliferation, (2) metabolic pathways maintaining stem cell quiescence, (3) metabolic control of cellular stress responses and cell death, (4) metabolic regulation of stem cell identity, and (5) metabolic requirements of the stem cell niche.


Stem Cells , Cell Differentiation/physiology , Cell Division
3.
Nat Metab ; 6(1): 127-140, 2024 Jan.
Article En | MEDLINE | ID: mdl-38172382

Mammalian preimplantation development is associated with marked metabolic robustness, and embryos can develop under a wide variety of nutrient conditions, including even the complete absence of soluble amino acids. Here we show that mouse embryonic stem cells (ESCs) capture the unique metabolic state of preimplantation embryos and proliferate in the absence of several essential amino acids. Amino acid independence is enabled by constitutive uptake of exogenous protein through macropinocytosis, alongside a robust lysosomal digestive system. Following transition to more committed states, ESCs reduce digestion of extracellular protein and instead become reliant on exogenous amino acids. Accordingly, amino acid withdrawal selects for ESCs that mimic the preimplantation epiblast. More broadly, we find that all lineages of preimplantation blastocysts exhibit constitutive macropinocytic protein uptake and digestion. Taken together, these results highlight exogenous protein uptake and digestion as an intrinsic feature of preimplantation development and provide insight into the catabolic strategies that enable embryos to sustain viability before implantation.


Blastocyst , Embryonic Stem Cells , Mice , Animals , Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Amino Acids/metabolism , Mammals/metabolism
5.
Cell ; 186(8): 1670-1688, 2023 04 13.
Article En | MEDLINE | ID: mdl-36858045

The uptake and metabolism of nutrients support fundamental cellular process from bioenergetics to biomass production and cell fate regulation. While many studies of cell metabolism focus on cancer cells, the principles of metabolism elucidated in cancer cells apply to a wide range of mammalian cells. The goal of this review is to discuss how the field of cancer metabolism provides a framework for revealing principles of cell metabolism and for dissecting the metabolic networks that allow cells to meet their specific demands. Understanding context-specific metabolic preferences and liabilities will unlock new approaches to target cancer cells to improve patient care.


Cells , Metabolic Networks and Pathways , Neoplasms , Animals , Humans , Cell Physiological Phenomena , Energy Metabolism , Mammals , Neoplasms/metabolism , Cells/metabolism
6.
Nat Rev Endocrinol ; 19(3): 134-150, 2023 03.
Article En | MEDLINE | ID: mdl-36446897

Tumours exhibit notable metabolic alterations compared with their corresponding normal tissue counterparts. These metabolic alterations can support anabolic growth, enable survival in hostile environments and regulate gene expression programmes that promote malignant progression. Whether these metabolic changes are selected for during malignant transformation or can themselves be drivers of tumour initiation is unclear. However, intriguingly, many of the major bottlenecks for tumour initiation - control of cell fate, survival and proliferation - are all amenable to metabolic regulation. In this article, we review evidence demonstrating a critical role for metabolic pathways in processes that support the earliest stages of tumour development. We discuss how cell-intrinsic factors, such as the cell of origin or transforming oncogene, and cell-extrinsic factors, such as local nutrient availability, promote or restrain tumour initiation. Deeper insight into how metabolic pathways control tumour initiation will improve our ability to design metabolic interventions to limit tumour incidence.


Neoplasms , Humans , Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Differentiation , Metabolic Networks and Pathways
7.
J Biol Chem ; 299(2): 102838, 2023 02.
Article En | MEDLINE | ID: mdl-36581208

The tricarboxylic acid (TCA) cycle, otherwise known as the Krebs cycle, is a central metabolic pathway that performs the essential function of oxidizing nutrients to support cellular bioenergetics. More recently, it has become evident that TCA cycle behavior is dynamic, and products of the TCA cycle can be co-opted in cancer and other pathologic states. In this review, we revisit the TCA cycle, including its potential origins and the history of its discovery. We provide a detailed accounting of the requirements for sustained TCA cycle function and the critical regulatory nodes that can stimulate or constrain TCA cycle activity. We also discuss recent advances in our understanding of the flexibility of TCA cycle wiring and the increasingly appreciated heterogeneity in TCA cycle activity exhibited by mammalian cells. Deeper insight into how the TCA cycle can be differentially regulated and, consequently, configured in different contexts will shed light on how this pathway is primed to meet the requirements of distinct mammalian cell states.


Citric Acid Cycle , Energy Metabolism , Animals , Citric Acid Cycle/physiology , Mammals
8.
Cell Stem Cell ; 29(12): 1621-1623, 2022 12 01.
Article En | MEDLINE | ID: mdl-36459965

As the stem cell community mourns the loss of New York Stem Cell Foundation founder Susan Solomon, we also look to celebrate her legacy. In this Voices, members of the 2022 class of NYSCF Roberston Investigators share how NYSCF community support will impact them and the bold ideas they will pursue as a result.


Research Personnel , Stem Cells , Female , Humans , New York , Community Support
10.
Nature ; 603(7901): 477-481, 2022 03.
Article En | MEDLINE | ID: mdl-35264789

The tricarboxylic acid (TCA) cycle is a central hub of cellular metabolism, oxidizing nutrients to generate reducing equivalents for energy production and critical metabolites for biosynthetic reactions. Despite the importance of the products of the TCA cycle for cell viability and proliferation, mammalian cells display diversity in TCA-cycle activity1,2. How this diversity is achieved, and whether it is critical for establishing cell fate, remains poorly understood. Here we identify a non-canonical TCA cycle that is required for changes in cell state. Genetic co-essentiality mapping revealed a cluster of genes that is sufficient to compose a biochemical alternative to the canonical TCA cycle, wherein mitochondrially derived citrate exported to the cytoplasm is metabolized by ATP citrate lyase, ultimately regenerating mitochondrial oxaloacetate to complete this non-canonical TCA cycle. Manipulating the expression of ATP citrate lyase or the canonical TCA-cycle enzyme aconitase 2 in mouse myoblasts and embryonic stem cells revealed that changes in the configuration of the TCA cycle accompany cell fate transitions. During exit from pluripotency, embryonic stem cells switch from canonical to non-canonical TCA-cycle metabolism. Accordingly, blocking the non-canonical TCA cycle prevents cells from exiting pluripotency. These results establish a context-dependent alternative to the traditional TCA cycle and reveal that appropriate TCA-cycle engagement is required for changes in cell state.


ATP Citrate (pro-S)-Lyase , Cell Differentiation , Citric Acid Cycle , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Animals , Citric Acid/metabolism , Embryonic Stem Cells , Mammals/metabolism , Mice , Mitochondria/metabolism , Pluripotent Stem Cells
11.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article En | MEDLINE | ID: mdl-35105808

Cells acquire essential nutrients from the environment and utilize adaptive mechanisms to survive when nutrients are scarce. How nutrients are trafficked and compartmentalized within cells and whether they are stored in response to stress remain poorly understood. Here, we investigate amino acid trafficking and uncover evidence for the lysosomal transit of numerous essential amino acids. We find that starvation induces the lysosomal retention of leucine in a manner requiring RAG-GTPases and the lysosomal protein complex Ragulator, but that this process occurs independently of mechanistic target of rapamycin complex 1 activity. We further find that stored leucine is utilized in protein synthesis and that inhibition of protein synthesis releases lysosomal stores. These findings identify a regulated starvation response that involves the lysosomal storage of leucine.


Leucine/metabolism , Lysosomes/metabolism , Signal Transduction , Stress, Physiological , Animals , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , RAW 264.7 Cells
13.
Nat Chem Biol ; 18(2): 180-190, 2022 02.
Article En | MEDLINE | ID: mdl-34937909

Fluorogenic RNA aptamers are used to genetically encode fluorescent RNA and to construct RNA-based metabolite sensors. Unlike naturally occurring aptamers that efficiently fold and undergo metabolite-induced conformational changes, fluorogenic aptamers can exhibit poor folding, which limits their cellular fluorescence. To overcome this, we evolved a naturally occurring well-folded adenine riboswitch into a fluorogenic aptamer. We generated a library of roughly 1015 adenine aptamer-like RNAs in which the adenine-binding pocket was randomized for both size and sequence, and selected Squash, which binds and activates the fluorescence of green fluorescent protein-like fluorophores. Squash exhibits markedly improved in-cell folding and highly efficient metabolite-dependent folding when fused to a S-adenosylmethionine (SAM)-binding aptamer. A Squash-based ratiometric sensor achieved quantitative SAM measurements, revealed cell-to-cell heterogeneity in SAM levels and revealed metabolic origins of SAM. These studies show that the efficient folding of naturally occurring aptamers can be exploited to engineer well-folded cell-compatible fluorogenic aptamers and devices.


Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Optical Imaging , Animals , Cells, Cultured , Embryo, Mammalian , Embryonic Stem Cells , Fluorescent Dyes/chemistry , Humans , Male , Mice , Nucleic Acid Conformation
14.
Science ; 374(6572): 1196-1197, 2021 Dec 03.
Article En | MEDLINE | ID: mdl-34855498

Fumarate siphons electrons to keep metabolism running.


Respiration
15.
Mol Cell ; 81(18): 3878-3878.e1, 2021 09 16.
Article En | MEDLINE | ID: mdl-34547243

Metabolic networks support cancer cell survival, proliferation, and malignant progression. Cancer cells take up large amounts of nutrients such as glucose and glutamine whose metabolism provides the energy, reducing equivalents, and biosynthetic precursors required to meet the biosynthetic demands of proliferation. Intermediates of glycolysis and the tricarboxylic acid (TCA) cycle provide critical building blocks for synthesis of non-essential amino acids, nucleotides, and fatty acids. To view this SnapShot, open or download the PDF.


Metabolic Networks and Pathways/physiology , Neoplasms/metabolism , Amino Acids/metabolism , Citric Acid Cycle/physiology , Energy Metabolism , Glucose/metabolism , Glutamine/metabolism , Glycolysis/physiology , Humans , Nucleotides/metabolism
16.
Ann N Y Acad Sci ; 1506(1): 55-73, 2021 12.
Article En | MEDLINE | ID: mdl-34414571

There is an increasing appreciation for the role of metabolism in cell signaling and cell decision making. Precise metabolic control is essential in development, as evident by the disorders caused by mutations in metabolic enzymes. The metabolic profile of cells is often cell-type specific, changing as cells differentiate or during tumorigenesis. Recent evidence has shown that changes in metabolism are not merely a consequence of changes in cell state but that metabolites can serve to promote and/or inhibit these changes. Metabolites can link metabolic pathways with cell signaling pathways via several mechanisms, for example, by serving as substrates for protein post-translational modifications, by affecting enzyme activity via allosteric mechanisms, or by altering epigenetic markers. Unraveling the complex interactions governing metabolism, gene expression, and protein activity that ultimately govern a cell's fate will require new tools and interactions across disciplines. On March 24 and 25, 2021, experts in cell metabolism, developmental biology, and human disease met virtually for the Keystone eSymposium, "Metabolic Decisions in Development and Disease." The discussions explored how metabolites impact cellular and developmental decisions in a diverse range of model systems used to investigate normal development, developmental disorders, dietary effects, and cancer-mediated changes in metabolism.


Congresses as Topic/trends , Human Development/physiology , Metabolic Diseases/physiopathology , Metabolic Networks and Pathways/physiology , Neoplasms/physiopathology , Research Report , Animals , Epigenesis, Genetic/physiology , Humans , Metabolic Diseases/genetics , Neoplasms/genetics , Signal Transduction/physiology
17.
Trends Cell Biol ; 31(1): 24-36, 2021 01.
Article En | MEDLINE | ID: mdl-33092942

Cell fate determination requires faithful execution of gene expression programs, which are increasingly recognized to respond to metabolic inputs. In particular, the family of α-ketoglutarate (αKG)-dependent dioxygenases, which include several chromatin-modifying enzymes, are emerging as key mediators of metabolic control of cell fate. αKG-dependent dioxygenases consume the metabolite αKG (also known as 2-oxoglutarate) as an obligate cosubstrate and are inhibited by succinate, fumarate, and 2-hydroxyglutarate. Here, we review the role of these metabolites in the control of dioxygenase activity and cell fate programs. We discuss the biochemical and transcriptional mechanisms enabling these metabolites to control cell fate and review evidence that nutrient availability shapes tissue-specific fate programs via αKG-dependent dioxygenases.


Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Cell Lineage , Animals , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Humans , Mutation/genetics , Neoplasms/enzymology , Neoplasms/genetics , Transcription Factors/metabolism
18.
Nat Cell Biol ; 22(11): 1396, 2020 Nov.
Article En | MEDLINE | ID: mdl-33046885

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

19.
Nat Cell Biol ; 22(7): 779-790, 2020 07.
Article En | MEDLINE | ID: mdl-32451440

Tissue stem cells are the cell of origin for many malignancies. Metabolites regulate the balance between self-renewal and differentiation, but whether endogenous metabolic pathways or nutrient availability predispose stem cells towards transformation remains unknown. Here, we address this question in epidermal stem cells (EpdSCs), which are a cell of origin for squamous cell carcinoma. We find that oncogenic EpdSCs are serine auxotrophs whose growth and self-renewal require abundant exogenous serine. When extracellular serine is limited, EpdSCs activate de novo serine synthesis, which in turn stimulates α-ketoglutarate-dependent dioxygenases that remove the repressive histone modification H3K27me3 and activate differentiation programmes. Accordingly, serine starvation or enforced α-ketoglutarate production antagonizes squamous cell carcinoma growth. Conversely, blocking serine synthesis or repressing α-ketoglutarate-driven demethylation facilitates malignant progression. Together, these findings reveal that extracellular serine is a critical determinant of EpdSC fate and provide insight into how nutrient availability is integrated with stem cell fate decisions during tumour initiation.


Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/pathology , Epidermal Cells/pathology , Ketoglutaric Acids/metabolism , Serine/metabolism , Stem Cells/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Epidermal Cells/metabolism , Female , Humans , Male , Mice , Stem Cells/metabolism
20.
Nat Metab ; 1(7): 676-687, 2019 07.
Article En | MEDLINE | ID: mdl-31511848

Most rapidly proliferating mammalian cells rely on the oxidation of exogenous glutamine to support cell proliferation. We previously found that culture of mouse embryonic stem cells (ESCs) in the presence of inhibitors against MEK and GSK3ß to maintain pluripotency reduces cellular reliance on glutamine for tricarboxylic acid (TCA) cycle anaplerosis, enabling ESCs to proliferate in the absence of exogenous glutamine. Here we show that reduced dependence on exogenous glutamine is a generalizable feature of pluripotent stem cells. Enhancing self-renewal, through either overexpression of pluripotency-associated transcription factors or altered signal transduction, decreases the utilization of glutamine-derived carbons in the TCA cycle. As a result, cells with the highest potential for self-renewal can be enriched by transient culture in glutamine-deficient media. During pluripotent cell culture or reprogramming to pluripotency, transient glutamine withdrawal selectively leads to the elimination of non-pluripotent cells. These data reveal that reduced dependence on glutamine anaplerosis is an inherent feature of self-renewing pluripotent stem cells and reveal a simple, non-invasive mechanism to select for mouse and human pluripotent stem cells within a heterogeneous population during both ESC passage and induced pluripotent cell reprogramming.


Glutamine/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cellular Reprogramming , Humans , Mice , Mouse Embryonic Stem Cells/metabolism
...