Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 356
Filter
1.
Nature ; 630(8018): 968-975, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867043

ABSTRACT

Obesity is a leading risk factor for progression and metastasis of many cancers1,2, yet can in some cases enhance survival3-5 and responses to immune checkpoint blockade therapies, including anti-PD-1, which targets PD-1 (encoded by PDCD1), an inhibitory receptor expressed on immune cells6-8. Although obesity promotes chronic inflammation, the role of the immune system in the obesity-cancer connection and immunotherapy remains unclear. It has been shown that in addition to T cells, macrophages can express PD-19-12. Here we found that obesity selectively induced PD-1 expression on tumour-associated macrophages (TAMs). Type I inflammatory cytokines and molecules linked to obesity, including interferon-γ, tumour necrosis factor, leptin, insulin and palmitate, induced macrophage PD-1 expression in an mTORC1- and glycolysis-dependent manner. PD-1 then provided negative feedback to TAMs that suppressed glycolysis, phagocytosis and T cell stimulatory potential. Conversely, PD-1 blockade increased the level of macrophage glycolysis, which was essential for PD-1 inhibition to augment TAM expression of CD86 and major histocompatibility complex I and II molecules and ability to activate T cells. Myeloid-specific PD-1 deficiency slowed tumour growth, enhanced TAM glycolysis and antigen-presentation capability, and led to increased CD8+ T cell activity with a reduced level of markers of exhaustion. These findings show that obesity-associated metabolic signalling and inflammatory cues cause TAMs to induce PD-1 expression, which then drives a TAM-specific feedback mechanism that impairs tumour immune surveillance. This may contribute to increased cancer risk yet improved response to PD-1 immunotherapy in obesity.


Subject(s)
Neoplasms , Obesity , Programmed Cell Death 1 Receptor , Tumor-Associated Macrophages , Animals , Female , Humans , Male , Mice , Antigen Presentation/drug effects , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Glycolysis/drug effects , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Obesity/immunology , Obesity/metabolism , Phagocytosis/drug effects , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects
2.
Cell Physiol Biochem ; 58(3): 250-272, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38865588

ABSTRACT

BACKGROUND/AIMS: Motivated by the vacuolar proton pump's importance in cancer, we investigate the effects of proton pump inhibition on breast cancer cell migration and proliferation, F-actin polymerization, lamin A/C, heterochromatin, and ETV7 expressions, nuclear size and shape, and AKT/mTOR signaling. METHODS: Lowly metastatic MCF7 and highly metastatic MDA-MB-231 breast cancer cells were treated with 120 nM of proton pump inhibitor Bafilomycin A1 for 24 hours. Cell migration was studied with wound- scratch assays, ATP levels with a chemiluminescent assay; cell proliferation was quantified by a cell area expansion assay. Nuclear size and shape were determined using DAPI nuclear stain and fluorescence microscopy. The levels of F-actin, lamin A/C, heterochromatin, and ETV7 were quantified using both immunocytochemistry and western blots; p-mTORC1, p-mTORC2, mTOR, p-AKT, and AKT were measured by western blots. RESULTS: We reveal that proton pump inhibition reduces F-actin polymerization, cell migration, proliferation, and increases heterochromatin in both lowly and highly metastatic cells. Surprisingly, Bafilomycin decreases lamin A/C in both cell lines. Inhibition has different effects on ETV7 expression in lowly and highly metastatic cells, as well as nuclear area, perimeter, and circularity. Bafilomycin also significantly decreases p-mTORC1, p-MTORC2, and MTOR expression in both cell lines, whereas it significantly decreases p-AKT in lowly metastatic cells and surprisingly significantly increases p-AKT in highly metastatic cells. Our proton pump inhibition protocol reduces V-ATPase levels (~25%) within three hours. V-ATPase levels vary in time for both control and inhibited cells, and inhibition reduces cellular ATP. CONCLUSION: Proton pumps promote F-actin polymerization and decrease heterochromatin, facilitating invasion. These pumps also upregulate both mTORC1 and mTORC2, thus highlighting the relevance of vacuolar proton pumps as metastatic cancer targets.


Subject(s)
Actins , Breast Neoplasms , Cell Movement , Cell Proliferation , Heterochromatin , Macrolides , Mechanistic Target of Rapamycin Complex 2 , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Vacuolar Proton-Translocating ATPases , Humans , Actins/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Signal Transduction/drug effects , Cell Movement/drug effects , Cell Line, Tumor , Female , Mechanistic Target of Rapamycin Complex 2/metabolism , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Macrolides/pharmacology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Cell Proliferation/drug effects , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Heterochromatin/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , MCF-7 Cells
3.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928292

ABSTRACT

Tanshinone IIA (T2A) is a bioactive compound that provides promise in the treatment of glioblastoma multiforme (GBM), with a range of molecular mechanisms including the inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy. Recently, T2A has been demonstrated to function through sestrin 2 (SESN) to inhibit mTORC1 activity, but its possible impact on autophagy through this pathway has not been investigated. Here, the model system Dictyostelium discoideum and GBM cell lines were employed to investigate the cellular role of T2A in regulating SESN to inhibit mTORC1 and activate autophagy through a GATOR2 component MIOS. In D. discoideum, T2A treatment induced autophagy and inhibited mTORC1 activity, with both effects lost upon the ablation of SESN (sesn-) or MIOS (mios-). We further investigated the targeting of MIOS to reproduce this effect of T2A, where computational analysis identified 25 novel compounds predicted to strongly bind the human MIOS protein, with one compound (MIOS inhibitor 3; Mi3) reducing cell proliferation in two GBM cells. Furthermore, Mi3 specificity was demonstrated through the loss of potency in the D. discoideum mios- cells regarding cell proliferation and the induction of autophagy. In GBM cells, Mi3 treatment also reduced mTORC1 activity and induced autophagy. Thus, a potential T2A mimetic showing the inhibition of mTORC1 and induction of autophagy in GBM cells was identified.


Subject(s)
Abietanes , Autophagy , Dictyostelium , Glioblastoma , Mechanistic Target of Rapamycin Complex 1 , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Abietanes/pharmacology , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Autophagy/drug effects , Cell Line, Tumor , Dictyostelium/drug effects , Dictyostelium/metabolism , Cell Proliferation/drug effects , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Sestrins
4.
Exp Eye Res ; 244: 109927, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750784

ABSTRACT

Subconjunctival fibrosis is the major cause of failure in both conventional and modern minimally invasive glaucoma surgeries (MIGSs) with subconjunctival filtration. The search for safe and effective anti-fibrotic agents is critical for improving long-term surgical outcomes. In this study, we investigated the effect of inhibiting the rapamycin-insensitive mTORC1/4E-BP1 axis on the transforming growth factor-beta 1(TGF-ß1)-induced fibrotic responses in human Tenon's fibroblasts (HTFs), as well as in a rat model of glaucoma filtration surgery (GFS). Primary cultured HTFs were treated with 3 ng/mL TGF-ß1 for 24 h, followed by treatment with 10 µM CZ415 for additional 24 h. Rapamycin (10 µM) was utilized as a control for mTORC1/4E-BP1 signaling insensitivity. The expression levels of fibrosis-associated molecules were measured using quantitative real-time PCR, Western blotting, and immunofluorescence analysis. Cell migration was assessed through the scratch wound assay. Additionally, a rat model of GFS was employed to evaluate the anti-fibrotic effect of CZ415 in vivo. Our findings indicated that both rapamycin and CZ415 treatment significantly reduced the TGF-ß1-induced cell proliferation, migration, and the expression of pro-fibrotic factors in HTFs. CZ415 also more effectively inhibited TGF-ß1-mediated collagen synthesis in HTFs compared to rapamycin. Activation of mTORC1/4E-BP signaling following TGF-ß1 exposure was highly suppressed by CZ415 but was only modestly inhibited by rapamycin. Furthermore, CZ415 was found to decrease subconjunctival collagen deposition in rats post GFS. Our results suggest that rapamycin-insensitive mTORC1/4E-BP1 signaling plays a critical role in TGF-ß1-driven collagen synthesis in HTFs. This study demonstrated that inhibition of the mTORC1/4E-BP1 axis offers superior anti-fibrotic efficacy compared to rapamycin and represents a promising target for improving the success rate of both traditional and modern GFSs.


Subject(s)
Adaptor Proteins, Signal Transducing , Fibroblasts , Fibrosis , Mechanistic Target of Rapamycin Complex 1 , Sirolimus , Tenon Capsule , Transforming Growth Factor beta1 , Animals , Transforming Growth Factor beta1/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Humans , Rats , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Sirolimus/pharmacology , Fibrosis/metabolism , Tenon Capsule/metabolism , Tenon Capsule/drug effects , Cells, Cultured , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Movement/drug effects , Disease Models, Animal , Blotting, Western , Rats, Sprague-Dawley , Cell Cycle Proteins/metabolism , Signal Transduction , Real-Time Polymerase Chain Reaction , Male , Glaucoma/metabolism , Glaucoma/drug therapy , Glaucoma/pathology , Immunosuppressive Agents/pharmacology
5.
Phytochemistry ; 222: 114105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657886

ABSTRACT

Three undescribed cassane diterpenoids, caesalpanins D-F (1-3), and seven known ones were isolated from the seeds of Caesalpinia sappan. Structures and absolute configurations of 1-3 were elucidated based on the extensive spectroscopic analysis, single-crystal X-ray diffraction analysis, and ECD calculations. Structurally, compound 1 was the first example of 18-norcassane diterpenoid and 2 was a rare 20-norcassane diterpenoid having an unusual five-membered oxygen bridge between C-10/C-18. The anti-proliferative activity of 1, 3, and 4-10 against PANC-1 cells (pancreatic ductal adenocarcinoma cell line) was evaluated, and phanginin H (4) was found to exhibit anti-cancer activity with IC50 value of 18.13 ± 0.63 µM. Compound 4 inhibited PANC-1 cell growth by arresting the cell cycle at G2/M phase via regulation of cyclin-dependent kinases, and the self-renewal and metastasis of PANC-1 cells by suppressing cancer cell stemness. Furthermore, compound 4 induced ROS generation and subsequently activated autophagy, which was demonstrated by the formation of autophagic vacuoles and dynamic change of autophagic flux. The induced ROS accumulation resulted in AMPK activation and subsequently regulation of mTORC1 activity and ULK phosphorylation, indicating that 4 triggered autophagy through ROS/AMPK/mTORC1 pathway. These findings suggested that 4 might potentially be an autophagy inducer for the therapy of pancreatic cancer.


Subject(s)
AMP-Activated Protein Kinases , Antineoplastic Agents, Phytogenic , Autophagy , Caesalpinia , Cell Proliferation , Diterpenes , Drug Screening Assays, Antitumor , Mechanistic Target of Rapamycin Complex 1 , Pancreatic Neoplasms , Reactive Oxygen Species , Seeds , Caesalpinia/chemistry , Humans , Diterpenes/pharmacology , Diterpenes/chemistry , Diterpenes/isolation & purification , Seeds/chemistry , Autophagy/drug effects , Reactive Oxygen Species/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/chemistry , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Cell Proliferation/drug effects , Molecular Structure , Cell Line, Tumor , Structure-Activity Relationship , Dose-Response Relationship, Drug
6.
Nat Commun ; 13(1): 5018, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028490

ABSTRACT

mTORC1 is hyperactive in multiple cancer types1,2. Here, we performed integrative analysis of single cell transcriptomic profiling, paired T cell receptor (TCR) sequencing, and spatial transcriptomic profiling on Tuberous Sclerosis Complex (TSC) associated tumors with mTORC1 hyperactivity, and identified a stem-like tumor cell state (SLS) linked to T cell dysfunction via tumor-modulated immunosuppressive macrophages. Rapamycin and its derivatives (rapalogs) are the primary treatments for TSC tumors, and the stem-like tumor cells showed rapamycin resistance in vitro, reminiscent of the cytostatic effects of these drugs in patients. The pro-angiogenic factor midkine (MDK) was highly expressed by the SLS population, and associated with enrichment of endothelial cells in SLS-dominant samples. Inhibition of MDK showed synergistic benefit with rapamycin in reducing the growth of TSC cell lines in vitro and in vivo. In aggregate, this study suggests an autocrine rapamycin resistance mechanism and a paracrine tumor survival mechanism via immune suppression adopted by the stem-like state tumor cells with mTORC1 hyperactivity.


Subject(s)
Mechanistic Target of Rapamycin Complex 1 , Midkine , Neoplasms , Neoplastic Stem Cells , Tumor Microenvironment , Endothelial Cells , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Midkine/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Sirolimus , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
7.
Nature ; 608(7922): 413-420, 2022 08.
Article in English | MEDLINE | ID: mdl-35922515

ABSTRACT

High cholesterol is a major risk factor for cardiovascular disease1. Currently, no drug lowers cholesterol through directly promoting cholesterol excretion. Human genetic studies have identified that the loss-of-function Asialoglycoprotein receptor 1 (ASGR1) variants associate with low cholesterol and a reduced risk of cardiovascular disease2. ASGR1 is exclusively expressed in liver and mediates internalization and lysosomal degradation of blood asialoglycoproteins3. The mechanism by which ASGR1 affects cholesterol metabolism is unknown. Here, we find that Asgr1 deficiency decreases lipid levels in serum and liver by stabilizing LXRα. LXRα upregulates ABCA1 and ABCG5/G8, which promotes cholesterol transport to high-density lipoprotein and excretion to bile and faeces4, respectively. ASGR1 deficiency blocks endocytosis and lysosomal degradation of glycoproteins, reduces amino-acid levels in lysosomes, and thereby inhibits mTORC1 and activates AMPK. On one hand, AMPK increases LXRα by decreasing its ubiquitin ligases BRCA1/BARD1. On the other hand, AMPK suppresses SREBP1 that controls lipogenesis. Anti-ASGR1 neutralizing antibody lowers lipid levels by increasing cholesterol excretion, and shows synergistic beneficial effects with atorvastatin or ezetimibe, two widely used hypocholesterolaemic drugs. In summary, this study demonstrates that targeting ASGR1 upregulates LXRα, ABCA1 and ABCG5/G8, inhibits SREBP1 and lipogenesis, and therefore promotes cholesterol excretion and decreases lipid levels.


Subject(s)
Asialoglycoprotein Receptor , Cholesterol , Lipid Metabolism , AMP-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Asialoglycoprotein Receptor/antagonists & inhibitors , Asialoglycoprotein Receptor/deficiency , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Asialoglycoproteins/metabolism , Atorvastatin/pharmacology , BRCA1 Protein , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cholesterol/metabolism , Drug Synergism , Endocytosis , Ezetimibe/pharmacology , Humans , Lipids/analysis , Lipids/blood , Liver/metabolism , Liver X Receptors/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1 , Ubiquitin-Protein Ligases/metabolism
8.
Science ; 376(6590): eabf8271, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35420934

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) remain without effective therapies. The mechanistic target of rapamycin complex 1 (mTORC1) pathway is a potential therapeutic target, but conflicting interpretations have been proposed for how mTORC1 controls lipid homeostasis. We show that selective inhibition of mTORC1 signaling in mice, through deletion of the RagC/D guanosine triphosphatase-activating protein folliculin (FLCN), promotes activation of transcription factor E3 (TFE3) in the liver without affecting other mTORC1 targets and protects against NAFLD and NASH. Disease protection is mediated by TFE3, which both induces lipid consumption and suppresses anabolic lipogenesis. TFE3 inhibits lipogenesis by suppressing proteolytic processing and activation of sterol regulatory element-binding protein-1c (SREBP-1c) and by interacting with SREBP-1c on chromatin. Our data reconcile previously conflicting studies and identify selective inhibition of mTORC1 as a potential approach to treat NASH and NAFLD.


Subject(s)
Mechanistic Target of Rapamycin Complex 1 , Non-alcoholic Fatty Liver Disease , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gene Deletion , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Non-alcoholic Fatty Liver Disease/therapy , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
9.
Cell Chem Biol ; 29(2): 259-275.e10, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34520743

ABSTRACT

Chemical splicing modulators that bind to the spliceosome have provided an attractive avenue for cancer treatment. Splicing modulators induce accumulation and subsequent translation of a subset of intron-retained mRNAs. However, the biological effect of proteins containing translated intron sequences remains unclear. Here, we identify a number of truncated proteins generated upon treatment with the splicing modulator spliceostatin A (SSA) via genome-wide ribosome profiling and bio-orthogonal noncanonical amino acid tagging (BONCAT) mass spectrometry. A subset of these truncated proteins has intrinsically disordered regions, forms insoluble cellular condensates, and triggers the proteotoxic stress response through c-Jun N-terminal kinase (JNK) phosphorylation, thereby inhibiting the mTORC1 pathway. In turn, this reduces global translation. These findings indicate that creating an overburden of condensate-prone proteins derived from introns represses translation and prevents further production of harmful truncated proteins. This mechanism appears to contribute to the antiproliferative and proapoptotic activity of splicing modulators.


Subject(s)
JNK Mitogen-Activated Protein Kinases/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , RNA Splicing/genetics , Spliceosomes/genetics , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Introns , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Pyrans/pharmacology , RNA Splicing/drug effects , RNA-Seq , Spiro Compounds/pharmacology , Spliceosomes/drug effects
10.
J Med Chem ; 65(2): 1329-1341, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34191518

ABSTRACT

Cancer-specific metabolic alterations hyperactivate the kinase activity of the mammalian/mechanistic target of rapamycin (mTOR) for overcoming stressful environments. Rapalogs, which allosterically inhibit mTOR complex 1 (mTORC1), have been approved as anticancer agents. However, the immunosuppressive side effect of these compounds results in the promotion of tumor metastasis, thereby limiting their therapeutic efficacy. We first report a nonrapalog inhibitor, WRX606, identified by a hybrid strategy of in silico and in cell selections. Our studies showed that WRX606 formed a ternary complex with FK506-binding protein-12 (FKBP12) and FKBP-rapamycin-binding (FRB) domain of mTOR, resulting in the allosteric inhibition of mTORC1. WRX606 inhibited the phosphorylation of not only the ribosomal protein S6 kinase 1 (S6K1) but also eIF4E-binding protein-1 (4E-BP1). Hence, WRX606 efficiently suppressed tumor growth in mice without promotion of metastasis. These results suggest that WRX606 is a potent lead compound for developing anticancer drugs discovered by in silico and in cell methods.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/drug therapy , Cell Cycle Proteins/metabolism , Computer Simulation , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Allosteric Regulation , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Proliferation , Female , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Nat Cell Biol ; 23(11): 1187-1198, 2021 11.
Article in English | MEDLINE | ID: mdl-34737445

ABSTRACT

How cancer cells adapt to evade the therapeutic effects of drugs targeting oncogenic drivers is poorly understood. Here we report an epigenetic mechanism leading to the adaptive resistance of triple-negative breast cancer (TNBC) to fibroblast growth factor receptor (FGFR) inhibitors. Prolonged FGFR inhibition suppresses the function of BRG1-dependent chromatin remodelling, leading to an epigenetic state that derepresses YAP-associated enhancers. These chromatin changes induce the expression of several amino acid transporters, resulting in increased intracellular levels of specific amino acids that reactivate mTORC1. Consistent with this mechanism, addition of mTORC1 or YAP inhibitors to FGFR blockade synergistically attenuated the growth of TNBC patient-derived xenograft models. Collectively, these findings reveal a feedback loop involving an epigenetic state transition and metabolic reprogramming that leads to adaptive therapeutic resistance and provides potential therapeutic strategies to overcome this mechanism of resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/metabolism , Drug Resistance, Neoplasm , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/drug therapy , YAP-Signaling Proteins/metabolism , Amino Acids/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Molecular Targeted Therapy , Multiprotein Complexes , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/genetics
12.
Science ; 374(6570): eabe7365, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34793210

ABSTRACT

Caloric restriction has been known for nearly a century to extend life span and delay age-associated pathology in laboratory animals. More recently, alternative "antiaging" diet modalities have been described that provide new mechanistic insights and potential clinical applications. These include intermittent fasting, fasting-mimicking diets, ketogenic diets, time-restricted feeding, protein restriction, and dietary restriction of specific amino acids. Despite mainstream popularization of some of these diets, many questions remain about their efficacy outside of a laboratory setting. Studies of these interventions support at least partially overlapping mechanisms of action and provide insights into what appear to be highly conserved mechanisms of biological aging.


Subject(s)
Aging , Diet , Health , Longevity , Amino Acids , Animals , Caloric Restriction/adverse effects , Diet/adverse effects , Diet Fads , Diet, Ketogenic/adverse effects , Diet, Protein-Restricted/adverse effects , Fasting/adverse effects , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
13.
Cells ; 10(10)2021 10 09.
Article in English | MEDLINE | ID: mdl-34685691

ABSTRACT

Seizure threshold 2 (SZT2) is a component of the KICSTOR complex which, under catabolic conditions, functions as a negative regulator in the amino acid-sensing branch of mTORC1. Mutations in this gene cause a severe neurodevelopmental and epileptic encephalopathy whose main symptoms include epilepsy, intellectual disability, and macrocephaly. As SZT2 remains one of the least characterized regulators of mTORC1, in this work we performed a systematic interactome analysis under catabolic and anabolic conditions. Besides numerous mTORC1 and AMPK signaling components, we identified clusters of proteins related to autophagy, ciliogenesis regulation, neurogenesis, and neurodegenerative processes. Moreover, analysis of SZT2 ablated cells revealed increased mTORC1 signaling activation that could be reversed by Rapamycin or Torin treatments. Strikingly, SZT2 KO cells also exhibited higher levels of autophagic components, independent of the physiological conditions tested. These results are consistent with our interactome data, in which we detected an enriched pool of selective autophagy receptors/regulators. Moreover, preliminary analyses indicated that SZT2 alters ciliogenesis. Overall, the data presented form the basis to comprehensively investigate the physiological functions of SZT2 that could explain major molecular events in the pathophysiology of developmental and epileptic encephalopathy in patients with SZT2 mutations.


Subject(s)
Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Protein Interaction Maps , Amino Acids/deficiency , Animals , Blood Proteins/pharmacology , Cilia/drug effects , Cilia/metabolism , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Organogenesis/drug effects , Principal Component Analysis , Protein Interaction Maps/drug effects , Sirolimus/pharmacology
14.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34622800

ABSTRACT

The role and mechanisms for upregulating complement factor B (CFB) expression in podocyte dysfunction in diabetic kidney disease (DKD) are not fully understood. Here, analyzing Gene Expression Omnibus GSE30528 data, we identified genes enriched in mTORC1 signaling, CFB, and complement alternative pathways in podocytes from patients with DKD. In mouse models, podocyte mTOR complex 1 (mTORC1) signaling activation was induced, while blockade of mTORC1 signaling reduced CFB upregulation, alternative complement pathway activation, and podocyte injury in the glomeruli. Knocking down CFB remarkably alleviated alternative complement pathway activation and DKD in diabetic mice. In cultured podocytes, high glucose treatment activated mTORC1 signaling, stimulated STAT1 phosphorylation, and upregulated CFB expression, while blockade of mTORC1 or STAT1 signaling abolished high glucose-upregulated CFB expression. Additionally, high glucose levels downregulated protein phosphatase 2Acα (PP2Acα) expression, while PP2Acα deficiency enhanced high glucose-induced mTORC1/STAT1 activation, CFB induction, and podocyte injury. Taken together, these findings uncover a mechanism by which CFB mediates podocyte injury in DKD.


Subject(s)
Complement Factor B/genetics , Diabetic Nephropathies/genetics , Hyperglycemia/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Podocytes/metabolism , STAT1 Transcription Factor/metabolism , Animals , Cells, Cultured , Complement Factor B/metabolism , Complement Pathway, Alternative , Databases, Genetic , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Gene Knockdown Techniques , Glucose/pharmacology , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Kidney/metabolism , Kidney/pathology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Podocytes/ultrastructure , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism , STAT1 Transcription Factor/antagonists & inhibitors
15.
Cancer Lett ; 523: 72-81, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34560229

ABSTRACT

Mechanistic target of rapamycin (mTOR) forms two distinct complexes, mTOR complex 1 (mTORC1) and mTORC2. Here we investigated the antitumor effect of dual mTORC1/2 inhibitor AZD2014 on epithelial ovarian cancer (EOC) and its potential effect on immunosuppressive myeloid-derived suppressor cells (MDSCs). Immunohistochemical analysis of mTORC1 and mTORC2 was performed on a human ovarian cancer tissue microarray. High mTORC2 expression level was associated with shorter survival in EOC, whereas mTORC1 was not correlate with patients' prognosis. AZD2014 suppressed mTOR signaling pathway in ovarian cancer cells, inhibited proliferation and induced G1-phase cell cycle arrest and apoptosis. In tumor-bearing mice, AZD2014 treatment limited tumor growth, reduced peritoneal ascites, and prolonged survival. AZD2014 specifically reduced MDSCs migration and accumulation in EOC peritoneal fluid but not in the spleen. Moreover, subsequent AZD2014 treatment after cisplatin chemotherapy delayed EOC recurrence. Collectively, we observed that high mTORC2 expression level in EOC indicated a poor prognosis. Remarkably, in tumor-bearing mice, AZD2014 diminished MDSC accumulation and delayed tumor growth and recurrence.


Subject(s)
Benzamides/pharmacology , Carcinoma, Ovarian Epithelial/drug therapy , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Morpholines/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Ovarian Neoplasms/drug therapy , Pyrimidines/pharmacology , Animals , Apoptosis , Benzamides/adverse effects , Benzamides/therapeutic use , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Female , Humans , Mechanistic Target of Rapamycin Complex 1/analysis , Mechanistic Target of Rapamycin Complex 2/analysis , Mice , Mice, Inbred C57BL , Morpholines/adverse effects , Morpholines/therapeutic use , Myeloid-Derived Suppressor Cells/physiology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , TOR Serine-Threonine Kinases/physiology , Xenograft Model Antitumor Assays
16.
Eur J Cancer ; 157: 268-277, 2021 11.
Article in English | MEDLINE | ID: mdl-34543871

ABSTRACT

AIM: Arms E and F of the AcSé-ESMART phase I/II platform trial aimed to define the recommended dose and preliminary activity of the dual mTORC1/2 inhibitor vistusertib as monotherapy and with topotecan-temozolomide in a molecularly enriched population of paediatric patients with relapsed/refractory malignancies. In addition, we evaluated genetic phosphatidylinositol 3-kinase (PI3K)/AKT/ mammalian (or mechanistic) target of rapamycin (mTOR) pathway alterations across the Molecular Profiling for Paediatric and Young Adult Cancer Treatment Stratification (MAPPYACTS) trial (NCT02613962). EXPERIMENTAL DESIGN AND RESULTS: Four patients were treated in arm E and 10 in arm F with a median age of 14.3 years. Main diagnoses were glioma and sarcoma. Dose escalation was performed as per the continuous reassessment method, expansion in an Ensign design. The vistusertib single agent administered at 75 mg/m2 twice a day (BID) on 2 days/week and vistusertib 30 mg/m2 BID on 3 days/week combined with temozolomide 100 mg/m2/day and topotecan 0.50 mg/m2/day on the first 5 days of each 4-week cycle were safe. Treatment was well tolerated with the main toxicity being haematological. Pharmacokinetics indicates equivalent exposure in children compared with adults. Neither tumour response nor prolonged stabilisation was observed, including in the 12 patients whose tumours exhibited PI3K/AKT/mTOR pathway alterations. Advanced profiling across relapsed/refractory paediatric cancers of the MAPPYACTS cohort shows genetic alterations associated with this pathway in 28.0% of patients, with 10.5% carrying mutations in the core pathway genes. CONCLUSIONS: Vistusertib was well tolerated in paediatric patients. Study arms were terminated because of the absence of tumour responses and insufficient target engagement of vistusertib observed in adult trials. Targeting the PI3K/AKT/mTOR pathway remains a therapeutic avenue to be explored in paediatric patients. CLINICAL TRIAL IDENTIFIER: NCT2813135.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides/administration & dosage , Morpholines/administration & dosage , Neoplasms/drug therapy , Pyrimidines/administration & dosage , Administration, Oral , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides/adverse effects , Child , Child, Preschool , Female , Gain of Function Mutation , Humans , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/genetics , Morpholines/adverse effects , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/genetics , Pyrimidines/adverse effects , Temozolomide/administration & dosage , Temozolomide/adverse effects , Topotecan/administration & dosage , Topotecan/adverse effects , Treatment Outcome , Young Adult
17.
Int J Oncol ; 59(4)2021 10.
Article in English | MEDLINE | ID: mdl-34523696

ABSTRACT

Mechanistic target of rapamycin (mTOR), which functions via two multiprotein complexes termed mTORC1 and mTORC2, is positioned in the canonical phosphoinositide 3­kinase­related kinase (PI3K)/AKT (PI3K/AKT) pathways. These complexes exert their actions by regulating other important kinases, such as 40S ribosomal S6 kinases (S6K), eukaryotic translation initiation factor 4E (elF4E)­binding protein 1 (4E­BP1) and AKT, to control cell growth, proliferation, migration and survival in response to nutrients and growth factors. Glioblastoma (GB) is a devastating form of brain cancer, where the mTOR pathway is deregulated due to frequent upregulation of the Receptor Tyrosine Kinase/PI3K pathways and loss of the tumor suppressor phosphatase and tensin homologue (PTEN). Rapamycin and its analogs were less successful in clinical trials for patients with GB due to their incomplete inhibition of mTORC1 and the activation of mitogenic pathways via negative feedback loops. Here, the effects of selective ATP­competitive dual inhibitors of mTORC1 and mTORC2, Torin1, Torin2 and XL388, are reported. Torin2 exhibited concentration­dependent pharmacodynamic effects on inhibition of phosphorylation of the mTORC1 substrates S6KSer235/236 and 4E­BP1Thr37/46 as well as the mTORC2 substrate AKTSer473 resulting in suppression of tumor cell migration, proliferation and S­phase entry. Torin1 demonstrated similar effects, but only at higher doses. XL388 suppressed cell proliferation at a higher dose, but failed to inhibit cell migration. Treatment with Torin1 suppressed phosphorylation of proline rich AKT substrate of 40 kDa (PRAS40) at Threonine 246 (PRAS40Thr246) whereas Torin2 completely abolished it. XL388 treatment suppressed the phosphorylation of PRAS40Thr246 only at higher doses. Drug resistance analysis revealed that treatment of GB cells with XL388 rendered partial drug resistance, which was also seen to a lesser extent with rapamycin and Torin1 treatments. However, treatment with Torin2 completely eradicated the tumor cell population. These results strongly suggest that Torin2, compared to Torin1 or XL388, is more effective in suppressing mTORC1 and mTORC2, and therefore in the inhibition of the GB cell proliferation, dissemination and in overcoming resistance to therapy. These findings underscore the significance of Torin2 in the treatment of GB.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , MTOR Inhibitors/pharmacology , Naphthyridines/pharmacology , Sulfones/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors
18.
Nat Commun ; 12(1): 4920, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389715

ABSTRACT

Malignant mesothelioma (MpM) is an aggressive, invariably fatal tumour that is causally linked with asbestos exposure. The disease primarily results from loss of tumour suppressor gene function and there are no 'druggable' driver oncogenes associated with MpM. To identify opportunities for management of this disease we have carried out polysome profiling to define the MpM translatome. We show that in MpM there is a selective increase in the translation of mRNAs encoding proteins required for ribosome assembly and mitochondrial biogenesis. This results in an enhanced rate of mRNA translation, abnormal mitochondrial morphology and oxygen consumption, and a reprogramming of metabolic outputs. These alterations delimit the cellular capacity for protein biosynthesis, accelerate growth and drive disease progression. Importantly, we show that inhibition of mRNA translation, particularly through combined pharmacological targeting of mTORC1 and 2, reverses these changes and inhibits malignant cell growth in vitro and in ex-vivo tumour tissue from patients with end-stage disease. Critically, we show that these pharmacological interventions prolong survival in animal models of asbestos-induced mesothelioma, providing the basis for a targeted, viable therapeutic option for patients with this incurable disease.


Subject(s)
Mesothelioma, Malignant/genetics , Oncogenes/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Animals , Asbestos , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/metabolism , Mesothelioma, Malignant/chemically induced , Mesothelioma, Malignant/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Naphthyridines/pharmacology , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Tumor Cells, Cultured
19.
FASEB J ; 35(9): e21788, 2021 09.
Article in English | MEDLINE | ID: mdl-34425031

ABSTRACT

Hypoxia increases fetal hepatic insulin-like growth factor binding protein-1 (IGFBP-1) phosphorylation mediated by mechanistic target of rapamycin (mTOR) inhibition. Whether maternal nutrient restriction (MNR) causes fetal hypoxia remains unclear. We used fetal liver from a baboon (Papio sp.) model of intrauterine growth restriction due to MNR (70% global diet of Control) and liver hepatocellular carcinoma (HepG2) cells as a model for human fetal hepatocytes and tested the hypothesis that mTOR-mediated IGFBP-1 hyperphosphorylation in response to hypoxia requires hypoxia-inducible factor-1α (HIF-1α) and regulated in development and DNA-damage responses-1 (REDD-1) signaling. Western blotting (n = 6) and immunohistochemistry (n = 3) using fetal liver indicated greater expression of HIF-1α, REDD-1 as well as erythropoietin and its receptor, and vascular endothelial growth factor at GD120 (GD185 term) in MNR versus Control. Moreover, treatment of HepG2 cells with hypoxia (1% pO2 ) (n = 3) induced REDD-1, inhibited mTOR complex-1 (mTORC1) activity and increased IGFBP-1 secretion/phosphorylation (Ser101/Ser119/Ser169). HIF-1α inhibition by echinomycin or small interfering RNA silencing prevented the hypoxia-mediated inhibition of mTORC1 and induction of IGFBP-1 secretion/phosphorylation. dimethyloxaloylglycine (DMOG) induced HIF-1α and also REDD-1 expression, inhibited mTORC1 and increased IGFBP-1 secretion/phosphorylation. Induction of HIF-1α (DMOG) and REDD-1 by Compound 3 inhibited mTORC1, increased IGFBP-1 secretion/ phosphorylation and protein kinase PKCα expression. Together, our data demonstrate that HIF-1α induction, increased REDD-1 expression and mTORC1 inhibition represent the mechanistic link between hypoxia and increased IGFBP-1 secretion/phosphorylation. We propose that maternal undernutrition limits fetal oxygen delivery, as demonstrated by increased fetal liver expression of hypoxia-responsive proteins in baboon MNR. These findings have important implications for our understanding of the pathophysiology of restricted fetal growth.


Subject(s)
Cell Culture Techniques , Disease Models, Animal , Fetal Growth Retardation/metabolism , Fetus/metabolism , Hypoxia/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Animals , Erythropoietin/metabolism , Fetal Weight , Fetus/chemistry , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Insulin-Like Growth Factor Binding Protein 1/chemistry , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Microscopy, Fluorescence , Organ Size , Papio , Phosphorylation , Protein Kinase C-alpha/metabolism , Receptors, Erythropoietin/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360912

ABSTRACT

Cellular senescence is a form of proliferative arrest triggered in response to a wide variety of stimuli and characterized by unique changes in cell morphology and function. Although unable to divide, senescent cells remain metabolically active and acquire the ability to produce and secrete bioactive molecules, some of which have recognized pro-inflammatory and/or pro-tumorigenic actions. As expected, this "senescence-associated secretory phenotype (SASP)" accounts for most of the non-cell-autonomous effects of senescent cells, which can be beneficial or detrimental for tissue homeostasis, depending on the context. It is now evident that many features linked to cellular senescence, including the SASP, reflect complex changes in the activities of mTOR and other metabolic pathways. Indeed, the available evidence indicates that mTOR-dependent signaling is required for the maintenance or implementation of different aspects of cellular senescence. Thus, depending on the cell type and biological context, inhibiting mTOR in cells undergoing senescence can reverse senescence, induce quiescence or cell death, or exacerbate some features of senescent cells while inhibiting others. Interestingly, autophagy-a highly regulated catabolic process-is also commonly upregulated in senescent cells. As mTOR activation leads to repression of autophagy in non-senescent cells (mTOR as an upstream regulator of autophagy), the upregulation of autophagy observed in senescent cells must take place in an mTOR-independent manner. Notably, there is evidence that autophagy provides free amino acids that feed the mTOR complex 1 (mTORC1), which in turn is required to initiate the synthesis of SASP components. Therefore, mTOR activation can follow the induction of autophagy in senescent cells (mTOR as a downstream effector of autophagy). These functional connections suggest the existence of autophagy regulatory pathways in senescent cells that differ from those activated in non-senescence contexts. We envision that untangling these functional connections will be key for the generation of combinatorial anti-cancer therapies involving pro-senescence drugs, mTOR inhibitors, and/or autophagy inhibitors.


Subject(s)
Autophagy , Cellular Senescence , Neoplasms/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cellular Senescence/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/drug therapy , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...