Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(36): eadh2023, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37672588

ABSTRACT

Previous studies have revealed a role for proline metabolism in supporting cancer development and metastasis. In this study, we show that many cancer cells respond to loss of attachment by accumulating and secreting proline. Detached cells display reduced proliferation accompanied by a general decrease in overall protein production and de novo amino acid synthesis compared to attached cells. However, proline synthesis was maintained under detached conditions. Furthermore, while overall proline incorporation into proteins was lower in detached cells compared to other amino acids, there was an increased production of the proline-rich protein collagen. The increased excretion of proline from detached cells was also shown to be used by macrophages, an abundant and important component of the tumor microenvironment. Our study suggests that detachment induced accumulation and secretion of proline may contribute to tumor progression by supporting increased production of extracellular matrix and providing proline to surrounding stromal cells.


Subject(s)
Neoplasms , Proline , Amino Acids , Biological Transport , Extracellular Matrix , Macrophages
2.
Cell Rep ; 42(6): 112562, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37245210

ABSTRACT

Mitochondrial 10-formyltetrahydrofolate (10-formyl-THF) is utilized by three mitochondrial enzymes to produce formate for nucleotide synthesis, NADPH for antioxidant defense, and formyl-methionine (fMet) to initiate mitochondrial mRNA translation. One of these enzymes-aldehyde dehydrogenase 1 family member 2 (ALDH1L2)-produces NADPH by catabolizing 10-formyl-THF into CO2 and THF. Using breast cancer cell lines, we show that reduction of ALDH1L2 expression increases ROS levels and the production of both formate and fMet. Both depletion of ALDH1L2 and direct exposure to formate result in enhanced cancer cell migration that is dependent on the expression of the formyl-peptide receptor (FPR). In various tumor models, increased ALDH1L2 expression lowers formate and fMet accumulation and limits metastatic capacity, while human breast cancer samples show a consistent reduction of ALDH1L2 expression in metastases. Together, our data suggest that loss of ALDH1L2 can support metastatic progression by promoting formate and fMet production, resulting in enhanced FPR-dependent signaling.


Subject(s)
Breast Neoplasms , Formates , Oxidoreductases Acting on CH-NH Group Donors , Female , Humans , Breast Neoplasms/metabolism , Formates/metabolism , Methionine , NADP , Reactive Oxygen Species , Oxidoreductases Acting on CH-NH Group Donors/metabolism
3.
Cell Metab ; 35(7): 1132-1146.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37230079

ABSTRACT

Augmented T cell function leading to host damage in autoimmunity is supported by metabolic dysregulation, making targeting immunometabolism an attractive therapeutic avenue. Canagliflozin, a type 2 diabetes drug, is a sodium glucose co-transporter 2 (SGLT2) inhibitor with known off-target effects on glutamate dehydrogenase and complex I. However, the effects of SGLT2 inhibitors on human T cell function have not been extensively explored. Here, we show that canagliflozin-treated T cells are compromised in their ability to activate, proliferate, and initiate effector functions. Canagliflozin inhibits T cell receptor signaling, impacting on ERK and mTORC1 activity, concomitantly associated with reduced c-Myc. Compromised c-Myc levels were encapsulated by a failure to engage translational machinery resulting in impaired metabolic protein and solute carrier production among others. Importantly, canagliflozin-treated T cells derived from patients with autoimmune disorders impaired their effector function. Taken together, our work highlights a potential therapeutic avenue for repurposing canagliflozin as an intervention for T cell-mediated autoimmunity.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 2 , Sodium-Glucose Transporter 2 Inhibitors , Humans , Canagliflozin/pharmacology , Canagliflozin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Autoimmunity , T-Lymphocytes , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Autoimmune Diseases/drug therapy , Hypoglycemic Agents/pharmacology
4.
Nature ; 615(7953): 705-711, 2023 03.
Article in English | MEDLINE | ID: mdl-36922598

ABSTRACT

Artificial sweeteners are used as calorie-free sugar substitutes in many food products and their consumption has increased substantially over the past years1. Although generally regarded as safe, some concerns have been raised about the long-term safety of the consumption of certain sweeteners2-5. In this study, we show that the intake of high doses of sucralose in mice results in immunomodulatory effects by limiting T cell proliferation and T cell differentiation. Mechanistically, sucralose affects the membrane order of T cells, accompanied by a reduced efficiency of T cell receptor signalling and intracellular calcium mobilization. Mice given sucralose show decreased CD8+ T cell antigen-specific responses in subcutaneous cancer models and bacterial infection models, and reduced T cell function in models of T cell-mediated autoimmunity. Overall, these findings suggest that a high intake of sucralose can dampen T cell-mediated responses, an effect that could be used in therapy to mitigate T cell-dependent autoimmune disorders.


Subject(s)
Sucrose , Sweetening Agents , T-Lymphocytes , Animals , Mice , Sucrose/analogs & derivatives , Sweetening Agents/administration & dosage , Sweetening Agents/adverse effects , Sweetening Agents/pharmacology , Sweetening Agents/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Food Safety , Calcium Signaling/drug effects , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/immunology , Bacterial Infections/immunology , Neoplasms/immunology , Autoimmunity/drug effects , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology
5.
Front Immunol ; 12: 637960, 2021.
Article in English | MEDLINE | ID: mdl-33868263

ABSTRACT

Regulatory T cells (Tregs) are essential for mitigating inflammation. Tregs are found in nearly every tissue and play either beneficial or harmful roles in the host. The availability of various nutrients can either enhance or impair Treg function. Mitochondrial oxidative metabolism plays a major role in supporting Treg differentiation and fitness. While Tregs rely heavily on oxidation of fatty acids to support mitochondrial activity, they have found ways to adapt to different tissue types, such as tumors, to survive in competitive environments. In addition, metabolic by-products from commensal organisms in the gut also have a profound impact on Treg differentiation. In this review, we will focus on the core metabolic pathways engaged in Tregs, especially in the context of tissue nutrient environments, and how they can affect Treg function, stability and differentiation.


Subject(s)
Inflammation/immunology , Microbiota/physiology , Mitochondria/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Humans , Immunomodulation , Nutrients , Oxidation-Reduction
6.
Nat Commun ; 12(1): 1209, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619282

ABSTRACT

Fructose intake has increased substantially throughout the developed world and is associated with obesity, type 2 diabetes and non-alcoholic fatty liver disease. Currently, our understanding of the metabolic and mechanistic implications for immune cells, such as monocytes and macrophages, exposed to elevated levels of dietary fructose is limited. Here, we show that fructose reprograms cellular metabolic pathways to favour glutaminolysis and oxidative metabolism, which are required to support increased inflammatory cytokine production in both LPS-treated human monocytes and mouse macrophages. A fructose-dependent increase in mTORC1 activity drives translation of pro-inflammatory cytokines in response to LPS. LPS-stimulated monocytes treated with fructose rely heavily on oxidative metabolism and have reduced flexibility in response to both glycolytic and mitochondrial inhibition, suggesting glycolysis and oxidative metabolism are inextricably coupled in these cells. The physiological implications of fructose exposure are demonstrated in a model of LPS-induced systemic inflammation, with mice exposed to fructose having increased levels of circulating IL-1ß after LPS challenge. Taken together, our work underpins a pro-inflammatory role for dietary fructose in LPS-stimulated mononuclear phagocytes which occurs at the expense of metabolic flexibility.


Subject(s)
Fructose/pharmacology , Glutamine/metabolism , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Acids/metabolism , Animals , Citric Acid Cycle/drug effects , Cytokines/metabolism , Disease Models, Animal , Glucose/pharmacology , Glycolysis/drug effects , Isotope Labeling , Macrophages/drug effects , Macrophages/metabolism , Metabolic Flux Analysis , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Monocytes/drug effects , Monocytes/metabolism , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Phenotype , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
7.
J Cell Sci ; 133(5)2020 03 06.
Article in English | MEDLINE | ID: mdl-32144194

ABSTRACT

The importance of cancer-cell-autonomous functions of the tumour suppressor p53 (encoded by TP53) has been established in many studies, but it is now clear that the p53 status of the cancer cell also has a profound impact on the immune response. Loss or mutation of p53 in cancers can affect the recruitment and activity of myeloid and T cells, allowing immune evasion and promoting cancer progression. p53 can also function in immune cells, resulting in various outcomes that can impede or support tumour development. Understanding the role of p53 in tumour and immune cells will help in the development of therapeutic approaches that can harness the differential p53 status of cancers compared with most normal tissue.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Immunity , Mutation , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
8.
Cell Rep ; 30(2): 481-496.e6, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940491

ABSTRACT

Loss of p53 function contributes to the development of many cancers. While cell-autonomous consequences of p53 mutation have been studied extensively, the role of p53 in regulating the anti-tumor immune response is still poorly understood. Here, we show that loss of p53 in cancer cells modulates the tumor-immune landscape to circumvent immune destruction. Deletion of p53 promotes the recruitment and instruction of suppressive myeloid CD11b+ cells, in part through increased expression of CXCR3/CCR2-associated chemokines and macrophage colony-stimulating factor (M-CSF), and attenuates the CD4+ T helper 1 (Th1) and CD8+ T cell responses in vivo. p53-null tumors also show an accumulation of suppressive regulatory T (Treg) cells. Finally, we show that two key drivers of tumorigenesis, activation of KRAS and deletion of p53, cooperate to promote immune tolerance.


Subject(s)
Myeloid Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Mice
9.
Cell Metab ; 30(4): 720-734.e5, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31447323

ABSTRACT

Cancer metastasis depends on cell survival following loss of extracellular matrix attachment and dissemination through the circulation. The metastatic spread can be enhanced by the clustering of detached cancer cells and increased antioxidant defense. Here, we link these responses by describing how cell clustering limits reactive oxygen species (ROS). Loss of attachment causes mitochondrial perturbations and increased ROS production. The formation of cell clusters induces a hypoxic environment that drives hypoxia-inducible factor 1-alpha (Hif1α)-mediated mitophagy, clearing damaged mitochondria and limiting ROS. However, hypoxia and reduced mitochondrial capacity promote dependence on glycolysis for ATP production that is supported by cytosolic reductive metabolism. Preventing this metabolic adaptation or disruption of cell clusters results in ROS accumulation, cell death, and a reduction of metastatic capacity in vivo. Our results provide a mechanistic explanation for the role of cell clustering in supporting survival during extracellular matrix detachment and metastatic spread and may point to targetable vulnerabilities.


Subject(s)
Mitochondria/metabolism , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Reactive Oxygen Species/metabolism , Animals , Cell Hypoxia , Cell Movement , Cell Survival , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Mitophagy
10.
Proc Natl Acad Sci U S A ; 116(18): 8975-8984, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30988188

ABSTRACT

T cell help in humoral immunity includes interactions of B cells with activated extrafollicular CD4+ and follicular T helper (Tfh) cells. Each can promote antibody responses but Tfh cells play critical roles during germinal center (GC) reactions. After restimulation of their antigen receptor (TCR) by B cells, helper T cells act on B cells via CD40 ligand and secreted cytokines that guide Ig class switching. Hypoxia is a normal feature of GC, raising questions about molecular mechanisms governing the relationship between hypoxia response mechanisms and T cell help to antibody responses. Hypoxia-inducible factors (HIF) are prominent among mechanisms that mediate cellular responses to limited oxygen but also are induced by lymphocyte activation. We now show that loss of HIF-1α or of both HIF-1α and HIF-2α in CD4+ T cells compromised essential functions in help during antibody responses. HIF-1α depletion from CD4+ T cells reduced frequencies of antigen-specific GC B cells, Tfh cells, and overall antigen-specific Ab after immunization with sheep red blood cells. Compound deficiency of HIF-1α and HIF-2α led to humoral defects after hapten-carrier immunization. Further, HIF promoted CD40L expression while restraining the FoxP3-positive CD4+ cells in the CXCR5+ follicular regulatory population. Glycolysis increases T helper cytokine expression, and HIF promoted glycolysis in T helper cells via TCR or cytokine stimulation, as well as their production of cytokines that direct antibody class switching. Indeed, IFN-γ elaboration by HIF-deficient in vivo-generated Tfh cells was impaired. Collectively, the results indicate that HIF transcription factors are vital components of the mechanisms of help during humoral responses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Antibody Formation , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Hypoxia/immunology , Cell Hypoxia/physiology , Cytokines/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Humans , Hypoxia/metabolism , Immunity, Humoral , Immunization , Lymphocyte Activation/immunology , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR5/metabolism , Sheep , T-Lymphocytes, Helper-Inducer/immunology
11.
Cell Metab ; 28(5): 721-736.e6, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30122553

ABSTRACT

Numerous mechanisms to support cells under conditions of transient nutrient starvation have been described. Several functions of the tumor-suppressor protein p53 can contribute to the adaptation of cells to metabolic stress and help cancer cell survival under nutrient-limiting conditions. We show here that p53 promotes the expression of SLC1A3, an aspartate/glutamate transporter that allows the utilization of aspartate to support cells in the absence of extracellular glutamine. Under glutamine deprivation, SLC1A3 expression maintains electron transport chain and tricarboxylic acid cycle activity, promoting de novo glutamate, glutamine, and nucleotide synthesis to rescue cell viability. Tumor cells with high levels of SLC1A3 expression are resistant to glutamine starvation, and SLC1A3 depletion retards the growth of these cells in vitro and in vivo, suggesting a therapeutic potential for SLC1A3 inhibition.


Subject(s)
Excitatory Amino Acid Transporter 1/metabolism , Glutamine/metabolism , Neoplasms/metabolism , Starvation/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptation, Physiological , Animals , Cell Line, Tumor , Cell Survival , Citric Acid Cycle , Female , Humans , Mice, Inbred BALB C
12.
Sci Immunol ; 3(23)2018 05 11.
Article in English | MEDLINE | ID: mdl-29752301

ABSTRACT

Mycobacterium tuberculosis (Mtb) is one of the most ancient human pathogens, yet the exact mechanism(s) of host defense against Mtb remains unclear. Although one-third of the world's population is chronically infected with Mtb, only 5 to 10% develop active disease. This indicates that, in addition to resistance mechanisms that control bacterial burden, the host has also evolved strategies to tolerate the presence of Mtb to limit disease severity. We identify mitochondrial cyclophilin D (CypD) as a critical checkpoint of T cell metabolism that controls the expansion of activated T cells. Although loss of CypD function in T cells led to enhanced Mtb antigen-specific T cell responses, this increased T cell response had no impact on bacterial burden. Rather, mice containing CypD-deficient T cells exhibited substantially compromised disease tolerance and succumbed to Mtb infection. This study establishes a mechanistic link between T cell-mediated immunity and disease tolerance during Mtb infection.


Subject(s)
Cyclophilins/immunology , Mitochondria/immunology , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/immunology , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis
13.
Proc Natl Acad Sci U S A ; 115(10): E2202-E2209, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463754

ABSTRACT

The translation of mRNAs into proteins serves as a critical regulatory event in gene expression. In the context of cancer, deregulated translation is a hallmark of transformation, promoting the proliferation, survival, and metastatic capabilities of cancer cells. The best-studied factor involved in the translational control of cancer is the eukaryotic translation initiation factor 4E (eIF4E). We and others have shown that eIF4E availability and phosphorylation promote metastasis in mouse models of breast cancer by selectively augmenting the translation of mRNAs involved in invasion and metastasis. However, the impact of translational control in cell types within the tumor microenvironment (TME) is unknown. Here, we demonstrate that regulatory events affecting translation in cells of the TME impact cancer progression. Mice bearing a mutation in the phosphorylation site of eIF4E (S209A) in cells comprising the TME are resistant to the formation of lung metastases in a syngeneic mammary tumor model. This is associated with reduced survival of prometastatic neutrophils due to decreased expression of the antiapoptotic proteins BCL2 and MCL1. Furthermore, we demonstrate that pharmacological inhibition of eIF4E phosphorylation prevents metastatic progression in vivo, supporting the development of phosphorylation inhibitors for clinical use.


Subject(s)
Breast Neoplasms/pathology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Neutrophils/metabolism , Protein Biosynthesis , Tumor Microenvironment , Amino Acid Motifs , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/chemistry , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Metastasis , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Nature ; 544(7650): 372-376, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28425994

ABSTRACT

The non-essential amino acids serine and glycine are used in multiple anabolic processes that support cancer cell growth and proliferation (reviewed in ref. 1). While some cancer cells upregulate de novo serine synthesis, many others rely on exogenous serine for optimal growth. Restriction of dietary serine and glycine can reduce tumour growth in xenograft and allograft models. Here we show that this observation translates into more clinically relevant autochthonous tumours in genetically engineered mouse models of intestinal cancer (driven by Apc inactivation) or lymphoma (driven by Myc activation). The increased survival following dietary restriction of serine and glycine in these models was further improved by antagonizing the anti-oxidant response. Disruption of mitochondrial oxidative phosphorylation (using biguanides) led to a complex response that could improve or impede the anti-tumour effect of serine and glycine starvation. Notably, Kras-driven mouse models of pancreatic and intestinal cancers were less responsive to depletion of serine and glycine, reflecting an ability of activated Kras to increase the expression of enzymes that are part of the serine synthesis pathway and thus promote de novo serine synthesis.


Subject(s)
Glycine/deficiency , Intestinal Neoplasms/diet therapy , Intestinal Neoplasms/metabolism , Lymphoma/diet therapy , Lymphoma/metabolism , Serine/deficiency , Animals , Antioxidants/metabolism , Biguanides/pharmacology , Cell Line, Tumor , Diet , Disease Models, Animal , Female , Food Deprivation , Glycine/metabolism , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Lymphoma/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nutritional Status , Oxidative Phosphorylation/drug effects , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Serine/biosynthesis , Serine/metabolism , Serine/pharmacology , Survival Rate
16.
Cancer Res ; 76(20): 6118-6129, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27530326

ABSTRACT

There is a need for transplantable murine models of ovarian high-grade serous carcinoma (HGSC) with regard to mutations in the human disease to assist investigations of the relationships between tumor genotype, chemotherapy response, and immune microenvironment. In addressing this need, we performed whole-exome sequencing of ID8, the most widely used transplantable model of ovarian cancer, covering 194,000 exomes at a mean depth of 400× with 90% exons sequenced >50×. We found no functional mutations in genes characteristic of HGSC (Trp53, Brca1, Brca2, Nf1, and Rb1), and p53 remained transcriptionally active. Homologous recombination in ID8 remained intact in functional assays. Further, we found no mutations typical of clear cell carcinoma (Arid1a, Pik3ca), low-grade serous carcinoma (Braf), endometrioid (Ctnnb1), or mucinous (Kras) carcinomas. Using CRISPR/Cas9 gene editing, we modeled HGSC by generating novel ID8 derivatives that harbored single (Trp53-/-) or double (Trp53-/-;Brca2-/-) suppressor gene deletions. In these mutants, loss of p53 alone was sufficient to increase the growth rate of orthotopic tumors with significant effects observed on the immune microenvironment. Specifically, p53 loss increased expression of the myeloid attractant CCL2 and promoted the infiltration of immunosuppressive myeloid cell populations into primary tumors and their ascites. In Trp53-/-;Brca2-/- mutant cells, we documented a relative increase in sensitivity to the PARP inhibitor rucaparib and slower orthotopic tumor growth compared with Trp53-/- cells, with an appearance of intratumoral tertiary lymphoid structures rich in CD3+ T cells. This work validates new CRISPR-generated models of HGSC to investigate its biology and promote mechanism-based therapeutics discovery. Cancer Res; 76(20); 6118-29. ©2016 AACR.


Subject(s)
BRCA2 Protein/physiology , CRISPR-Cas Systems/physiology , Cystadenocarcinoma, Serous/etiology , Disease Models, Animal , Ovarian Neoplasms/etiology , Tumor Suppressor Protein p53/physiology , Animals , BRCA2 Protein/genetics , Cell Line, Tumor , Cystadenocarcinoma, Serous/drug therapy , Exome , Female , Gene Editing , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics
17.
Mol Cell ; 60(2): 195-207, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26474064

ABSTRACT

Cancer cells adapt metabolically to proliferate under nutrient limitation. Here we used combined transcriptional-metabolomic network analysis to identify metabolic pathways that support glucose-independent tumor cell proliferation. We found that glucose deprivation stimulated re-wiring of the tricarboxylic acid (TCA) cycle and early steps of gluconeogenesis to promote glucose-independent cell proliferation. Glucose limitation promoted the production of phosphoenolpyruvate (PEP) from glutamine via the activity of mitochondrial PEP-carboxykinase (PCK2). Under these conditions, glutamine-derived PEP was used to fuel biosynthetic pathways normally sustained by glucose, including serine and purine biosynthesis. PCK2 expression was required to maintain tumor cell proliferation under limited-glucose conditions in vitro and tumor growth in vivo. Elevated PCK2 expression is observed in several human tumor types and enriched in tumor tissue from non-small-cell lung cancer (NSCLC) patients. Our results define a role for PCK2 in cancer cell metabolic reprogramming that promotes glucose-independent cell growth and metabolic stress resistance in human tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Gluconeogenesis/genetics , Lung Neoplasms/metabolism , Neoplasms/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Adaptation, Physiological/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle/genetics , Glucose/deficiency , Glutamine/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Metabolomics , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/pathology , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Purines/biosynthesis , Pyruvic Acid/metabolism , Serine/biosynthesis
18.
Elife ; 42015 Oct 23.
Article in English | MEDLINE | ID: mdl-26496200

ABSTRACT

T follicular helper cells (Tfh) are critical for the longevity and quality of antibody-mediated protection against infection. Yet few signaling pathways have been identified to be unique solely to Tfh development. ROQUIN is a post-transcriptional repressor of T cells, acting through its ROQ domain to destabilize mRNA targets important for Th1, Th17, and Tfh biology. Here, we report that ROQUIN has a paradoxical function on Tfh differentiation mediated by its RING domain: mice with a T cell-specific deletion of the ROQUIN RING domain have unchanged Th1, Th2, Th17, and Tregs during a T-dependent response but show a profoundly defective antigen-specific Tfh compartment. ROQUIN RING signaling directly antagonized the catalytic α1 subunit of adenosine monophosphate-activated protein kinase (AMPK), a central stress-responsive regulator of cellular metabolism and mTOR signaling, which is known to facilitate T-dependent humoral immunity. We therefore unexpectedly uncover a ROQUIN-AMPK metabolic signaling nexus essential for selectively promoting Tfh responses.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Differentiation , Signal Transduction , T-Lymphocytes, Helper-Inducer/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Mice , Sequence Deletion , Ubiquitin-Protein Ligases/genetics
19.
Cell Metab ; 22(4): 577-89, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26365179

ABSTRACT

Metabolic reprogramming is a hallmark of cellular transformation, yet little is known about metabolic changes that accompany tumor metastasis. Here we show that primary breast cancer cells display extensive metabolic heterogeneity and engage distinct metabolic programs depending on their site of metastasis. Liver-metastatic breast cancer cells exhibit a unique metabolic program compared to bone- or lung-metastatic cells, characterized by increased conversion of glucose-derived pyruvate into lactate and a concomitant reduction in mitochondrial metabolism. Liver-metastatic cells displayed increased HIF-1α activity and expression of the HIF-1α target Pyruvate dehydrogenase kinase-1 (PDK1). Silencing HIF-1α reversed the glycolytic phenotype of liver-metastatic cells, while PDK1 was specifically required for metabolic adaptation to nutrient limitation and hypoxia. Finally, we demonstrate that PDK1 is required for efficient liver metastasis, and its expression is elevated in liver metastases from breast cancer patients. Our data implicate PDK1 as a key regulator of metabolism and metastatic potential in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Metabolic Engineering , Protein Serine-Threonine Kinases/metabolism , Animals , Breast Neoplasms/metabolism , Carbon Isotopes/chemistry , Carbon Isotopes/metabolism , Cell Hypoxia , Cell Line, Tumor , Female , Gas Chromatography-Mass Spectrometry , Glutamine/metabolism , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Metabolome , Mice , Mice, Inbred BALB C , Oxidative Phosphorylation , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering/metabolism
20.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607458

ABSTRACT

Naive T cells undergo metabolic reprogramming to support the increased energetic and biosynthetic demands of effector T cell function. However, how nutrient availability influences T cell metabolism and function remains poorly understood. Here we report plasticity in effector T cell metabolism in response to changing nutrient availability. Activated T cells were found to possess a glucose-sensitive metabolic checkpoint controlled by the energy sensor AMP-activated protein kinase (AMPK) that regulated mRNA translation and glutamine-dependent mitochondrial metabolism to maintain T cell bioenergetics and viability. T cells lacking AMPKα1 displayed reduced mitochondrial bioenergetics and cellular ATP in response to glucose limitation in vitro or pathogenic challenge in vivo. Finally, we demonstrated that AMPKα1 is essential for T helper 1 (Th1) and Th17 cell development and primary T cell responses to viral and bacterial infections in vivo. Our data highlight AMPK-dependent regulation of metabolic homeostasis as a key regulator of T cell-mediated adaptive immunity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/metabolism , AMP-Activated Protein Kinases/genetics , Adaptation, Physiological/immunology , Animals , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Energy Metabolism , Glucose/metabolism , Glutamine/metabolism , Humans , Immunomodulation , Lymphocyte Activation/genetics , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Protein Biosynthesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...