Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cell ; 167(2): 405-418.e13, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693350

ABSTRACT

The HVEM (TNFRSF14) receptor gene is among the most frequently mutated genes in germinal center lymphomas. We report that loss of HVEM leads to cell-autonomous activation of B cell proliferation and drives the development of GC lymphomas in vivo. HVEM-deficient lymphoma B cells also induce a tumor-supportive microenvironment marked by exacerbated lymphoid stroma activation and increased recruitment of T follicular helper (TFH) cells. These changes result from the disruption of inhibitory cell-cell interactions between the HVEM and BTLA (B and T lymphocyte attenuator) receptors. Accordingly, administration of the HVEM ectodomain protein (solHVEM(P37-V202)) binds BTLA and restores tumor suppression. To deliver solHVEM to lymphomas in vivo, we engineered CD19-targeted chimeric antigen receptor (CAR) T cells that produce solHVEM locally and continuously. These modified CAR-T cells show enhanced therapeutic activity against xenografted lymphomas. Hence, the HVEM-BTLA axis opposes lymphoma development, and our study illustrates the use of CAR-T cells as "micro-pharmacies" able to deliver an anti-cancer protein.


Subject(s)
Adoptive Transfer/methods , Lymphoma, Follicular/therapy , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/genetics , T-Lymphocytes/immunology , Tumor Suppressor Proteins/genetics , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , Cell Proliferation , Humans , Lymphocyte Activation , Lymphoma, Follicular/genetics , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Protein Domains , Protein Engineering , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
2.
J Immunol ; 209(6): 1189-1199, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36002234

ABSTRACT

The activation of memory T cells is a very rapid and concerted cellular response that requires coordination between cellular processes in different compartments and on different time scales. In this study, we use ribosome profiling and deep RNA sequencing to define the acute mRNA translation changes in CD8 memory T cells following initial activation events. We find that initial translation enables subsequent events of human and mouse T cell activation and expansion. Briefly, early events in the activation of Ag-experienced CD8 T cells are insensitive to transcriptional blockade with actinomycin D, and instead depend on the translation of pre-existing mRNAs and are blocked by cycloheximide. Ribosome profiling identifies ∼92 mRNAs that are recruited into ribosomes following CD8 T cell stimulation. These mRNAs typically have structured GC and pyrimidine-rich 5' untranslated regions and they encode key regulators of T cell activation and proliferation such as Notch1, Ifngr1, Il2rb, and serine metabolism enzymes Psat1 and Shmt2 (serine hydroxymethyltransferase 2), as well as translation factors eEF1a1 (eukaryotic elongation factor α1) and eEF2 (eukaryotic elongation factor 2). The increased production of receptors of IL-2 and IFN-γ precedes the activation of gene expression and augments cellular signals and T cell activation. Taken together, we identify an early RNA translation program that acts in a feed-forward manner to enable the rapid and dramatic process of CD8 memory T cell expansion and activation.


Subject(s)
Glycine Hydroxymethyltransferase , Interleukin-2 , 5' Untranslated Regions , Animals , CD8-Positive T-Lymphocytes , Cycloheximide/metabolism , Dactinomycin/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Humans , Immunologic Memory , Interleukin-2/metabolism , Lymphocyte Activation , Memory T Cells , Mice , Peptide Elongation Factor 2/genetics , Peptide Elongation Factor 2/metabolism , Peptide Elongation Factors/genetics , Pyrimidines/metabolism , RNA, Messenger/genetics , Serine/genetics
3.
Proc Natl Acad Sci U S A ; 114(36): E7441-E7449, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827325

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma in adults. DLBCL exhibits highly aggressive and systemic progression into multiple tissues in patients, particularly in lymph nodes. Whole-body 18F-fluodeoxyglucose positron emission tomography ([18F]FDG-PET) imaging has an essential role in diagnosing DLBCL in the clinic; however, [18F]FDG-PET often faces difficulty in differentiating malignant tissues from certain nonmalignant tissues with high glucose uptake. We have developed a PET imaging strategy for DLBCL that targets poly[ADP ribose] polymerase 1 (PARP1), the expression of which has been found to be much higher in DLBCL than in healthy tissues. In a syngeneic DLBCL mouse model, this PARP1-targeted PET imaging approach allowed us to discriminate between malignant and inflamed lymph nodes, whereas [18F]FDG-PET failed to do so. Our PARP1-targeted PET imaging approach may be an attractive addition to the current PET imaging strategy to differentiate inflammation from malignancy in DLBCL.


Subject(s)
Lymph Nodes/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Animals , Cell Line, Tumor , Female , Fluorodeoxyglucose F18/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods , Radiopharmaceuticals/administration & dosage
4.
J Biol Chem ; 292(3): 936-944, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27956548

ABSTRACT

Lipids are important nutrients that proliferating cells require to maintain energy homeostasis as well as to build plasma membranes for newly synthesized cells. Previously, we identified nutrient-sensing checkpoints that exist in the latter part of the G1 phase of the cell cycle that are dependent upon essential amino acids, Gln, and finally, a checkpoint mediated by mammalian target of rapamycin (mTOR), which integrates signals from both nutrients and growth factors. In this study, we have identified and temporally mapped a lipid-mediated G1 checkpoint. This checkpoint is located after the Gln checkpoint and before the mTOR-mediated cell cycle checkpoint. Intriguingly, clear cell renal cell carcinoma cells (ccRCC) have a dysregulated lipid-mediated checkpoint due in part to defective phosphatase and tensin homologue (PTEN). When deprived of lipids, instead of arresting in G1, these cells continue to cycle and utilize lipid droplets as a source of lipids. Lipid droplets have been known to maintain endoplasmic reticulum homeostasis and prevent cytotoxic endoplasmic reticulum stress in ccRCC. Dysregulation of the lipid-mediated checkpoint forces these cells to utilize lipid droplets, which could potentially lead to therapeutic opportunities that exploit this property of ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Membrane/metabolism , G1 Phase Cell Cycle Checkpoints , Lipid Metabolism , Carcinoma, Renal Cell/pathology , Cell Membrane/pathology , Endoplasmic Reticulum Stress , Glutamine/metabolism , Humans , Kidney Neoplasms , MCF-7 Cells , Neoplasm Proteins/metabolism , PTEN Phosphohydrolase/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
J Biol Chem ; 292(15): 6303-6311, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28223357

ABSTRACT

mTOR, the mammalian target of rapamycin, integrates growth factor and nutrient signals to promote a transformation from catabolic to anabolic metabolism, cell growth, and cell cycle progression. Phosphatidic acid (PA) interacts with the FK506-binding protein-12-rapamycin-binding (FRB) domain of mTOR, which stabilizes both mTOR complexes: mTORC1 and mTORC2. We report here that mTORC1 and mTORC2 are activated in response to exogenously supplied fatty acids via the de novo synthesis of PA, a central metabolite for membrane phospholipid biosynthesis. We examined the impact of exogenously supplied fatty acids on mTOR in KRas-driven cancer cells, which are programmed to utilize exogenous lipids. The induction of mTOR by oleic acid was dependent upon the enzymes responsible for de novo synthesis of PA. Suppression of the de novo synthesis of PA resulted in G1 cell cycle arrest. Although it has long been appreciated that mTOR is a sensor of amino acids and glucose, this study reveals that mTOR also senses the presence of lipids via production of PA.


Subject(s)
Multiprotein Complexes/metabolism , Phosphatidic Acids/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , MCF-7 Cells , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Oleic Acid/pharmacology , Phosphatidic Acids/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , TOR Serine-Threonine Kinases/genetics
6.
J Biol Chem ; 289(33): 22583-22588, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24990952

ABSTRACT

Phosphatidic acid (PA) is a critical metabolite at the heart of membrane phospholipid biosynthesis. However, PA also serves as a critical lipid second messenger that regulates several proteins implicated in the control of cell cycle progression and cell growth. Three major metabolic pathways generate PA: phospholipase D (PLD), diacylglycerol kinase (DGK), and lysophosphatidic acid acyltransferase (LPAAT). The LPAAT pathway is integral to de novo membrane phospholipid biosynthesis, whereas the PLD and DGK pathways are activated in response to growth factors and stress. The PLD pathway is also responsive to nutrients. A key target for the lipid second messenger function of PA is mTOR, the mammalian/mechanistic target of rapamycin, which integrates both nutrient and growth factor signals to control cell growth and proliferation. Although PLD has been widely implicated in the generation of PA needed for mTOR activation, it is becoming clear that PA generated via the LPAAT and DGK pathways is also involved in the regulation of mTOR. In this minireview, we highlight the coordinated maintenance of intracellular PA levels that regulate mTOR signals stimulated by growth factors and nutrients, including amino acids, lipids, glucose, and Gln. Emerging evidence indicates compensatory increases in one source of PA when another source is compromised, highlighting the importance of being able to adapt to stressful conditions that interfere with PA production. The regulation of PA levels has important implications for cancer cells that depend on PA and mTOR activity for survival.


Subject(s)
Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Second Messenger Systems/physiology , TOR Serine-Threonine Kinases/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Animals , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Glucose/genetics , Glucose/metabolism , Glutamine/genetics , Glutamine/metabolism , Humans , Phosphatidic Acids/genetics , Phospholipase D/genetics , TOR Serine-Threonine Kinases/genetics
7.
J Biol Chem ; 286(29): 25477-86, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21622984

ABSTRACT

The mammalian target of rapamycin (mTOR) is a critical sensor of nutritional sufficiency. Although much is known about the regulation of mTOR in response to growth factors, much less is known about the regulation of mTOR in response to nutrients. Amino acids have no impact on the signals that regulate Rheb, a GTPase required for the activation of mTOR complex 1 (mTORC1). Phospholipase D (PLD) generates a metabolite, phosphatidic acid, that facilitates association between mTOR and the mTORC1 co-factor Raptor. We report here that elevated PLD activity in human cancer cells is dependent on both amino acids and glucose and that amino acid- and glucose-induced increases in mTORC1 activity are dependent on PLD. Amino acid- and glucose-induced PLD and mTORC1 activity were also dependent on the GTPases RalA and ARF6 and the type III phosphatidylinositol-3-kinase hVps34. Thus, a key stimulatory event for mTORC1 activation in response to nutrients is the generation of phosphatidic acid by PLD.


Subject(s)
Food , Phospholipase D/metabolism , Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Amino Acids/pharmacology , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/metabolism , Glucose/pharmacology , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Neuropeptides/metabolism , Phosphatidic Acids/metabolism , Ras Homolog Enriched in Brain Protein , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , ral GTP-Binding Proteins/metabolism
8.
Cancer Discov ; 8(8): 958-971, 2018 08.
Article in English | MEDLINE | ID: mdl-29880584

ABSTRACT

CD19-specific chimeric antigen receptor (CAR) T-cell therapy is highly effective against relapsed or refractory acute lymphoblastic leukemia (ALL), but is hindered by neurotoxicity. In 53 adult patients with ALL, we found a significant association of severe neurotoxicity with high pretreatment disease burden, higher peak CAR T-cell expansion, and early and higher elevations of proinflammatory cytokines in blood. Patients with severe neurotoxicity had evidence of blood-cerebrospinal fluid (CSF) barrier disruption correlating with neurotoxicity grade without association with CSF white blood cell count or CAR T-cell quantity in CSF. Proinflammatory cytokines were enriched in CSF during severe neurotoxicity with disproportionately high levels of IL6, IL8, MCP1, and IP10, suggesting central nervous system-specific production. Seizures, seizure-like activity, myoclonus, and neuroimaging characteristics suggested excitatory neurotoxicity, and we found elevated levels of endogenous excitatory agonists in CSF during neurotoxicity.Significance: We detail the neurologic symptoms and blood, CSF, and neuroimaging correlates of neurotoxicity associated with CD19 CAR T cells and identify neurotoxicity risk factors. Our findings implicate cellular components other than T cells and suggest novel links between systemic inflammation and characteristic neurotoxicity symptoms. Cancer Discov; 8(8); 958-71. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.


Subject(s)
Adoptive Transfer/adverse effects , Antigens, CD19/immunology , Cytokines/cerebrospinal fluid , Neurotoxicity Syndromes/diagnostic imaging , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/transplantation , Adrenal Cortex Hormones/therapeutic use , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Cytokines/blood , Female , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neuroimaging/methods , Neurotoxicity Syndromes/cerebrospinal fluid , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Tumor Burden , Young Adult
9.
Mol Cancer Ther ; 13(3): 733-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24435447

ABSTRACT

The conversion of normal cells to cancer cells involves a shift from catabolic to anabolic metabolism involving increased glucose uptake and the diversion of glycolytic intermediates into nucleotides, amino acids, and lipids needed for cell growth. An underappreciated aspect of nutrient uptake is the utilization of serum lipids. We investigated the dependence of human cancer cells on serum lipids and report here that Ras-driven human cancer cells are uniquely dependent on serum lipids for both proliferation and survival. Removal of serum lipids also sensitizes Ras-driven cancer cells to rapamycin-indicating that the enhanced need for serum lipids creates a synthetic lethal phenotype that could be exploited therapeutically. Although depriving humans of serum lipids is not practical, suppressing uptake of lipids is possible. Suppressing macropinocytosis in Ras-driven cancer cells also created sensitivity to suppression of the mammalian/mechanistic target of rapamycin complex 1 (mTORC1). It is speculated that this property displayed by Ras-driven cancer cells represents an Achilles' heel for the large number of human cancers that are driven by activating Ras mutations.


Subject(s)
Carcinogenesis/genetics , Lipids/genetics , Multiprotein Complexes/genetics , TOR Serine-Threonine Kinases/genetics , ras Proteins/genetics , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Lipids/administration & dosage , Lipids/blood , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Mutation , ras Proteins/blood
10.
Cell Cycle ; 10(22): 3948-56, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22071574

ABSTRACT

mTOR, the mammalian target of rapamycin, has been widely implicated in signals that promote cell cycle progression and survival in cancer cells. Rapamycin, which inhibits mTOR with high specificity, has consequently attracted much attention as an anti-cancer therapeutic. Rapamycin suppresses phosphorylation of S6 kinase at nano-molar concentrations, however at higher micro-molar doses, rapamycin induces apoptosis in several human cancer cell lines. While much is known about the effect of low dose rapamycin treatment, the mechanistic basis for the apoptotic effects of high-dose rapamycin treatment is not understood. We report here that the apoptotic effects of high-dose rapamycin treatment correlate with suppressing phosphorylation of the mTOR complex 1 substrate, eukaryotic initiation factor 4E (eIF4E) binding protein-1 (4E-BP1). Consistent with this observation, ablation of eIF4E also resulted in apoptorsis in MDA-MB 231 breast cancer cells. We also provide evidence that the differential dose effects of rapamycin are correlated with partial and complete dissociation of Raptor from mTORC1 at low and high doses, respectively. In contrast with MDA-MB-231 cells, MCF-7 breast cancer cells survived rapamycin-induced suppression of 4E-BP1 phosphorylation. We show that survival correlated with a hyper-phosphorylation of Akt at S473 at high rapamycin doses, the suppression of which conferred rapamycin sensitivity. This study reveals that the apoptotic effect of rapamycin requires doses that completely dissociate Raptor from mTORC1 and suppress that phosphorylation of 4E-BP1 and inhibit eIF4E.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Phosphoproteins/metabolism , Proteins/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology , Cell Cycle/drug effects , Cell Cycle Proteins , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL