Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
IEEE Trans Biomed Eng ; 71(4): 1104-1114, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37874730

ABSTRACT

OBJECTIVE: In the process of cochlear implantation surgery, it is crucial to develop a method to control the temperature during the drilling of the implant channel since high temperatures can result in damage to bone and nerve tissue. METHODS: This paper simplified the traditional point heat source temperature rise model and proposed a novel extreme peck drilling model to quantitatively calculate the maximum temperature rise value. It is also innovatively introduced a new method for calculating the best peck drilling duty cycle to strictly control the maximum temperature rise value. Besides, the neural network is trained with virtual data to identify two important thermal parameters in the temperature rise model. RESULTS: In the experiment of epoxy resin and temporal bone, the difference between predicted maximum temperature and actual maximum temperature was less than 1.5 °C, and the error rate was less than 10%. And the error source was analyzed by variational mode decomposition, along with discussion of potential solutions. In the temperature control experiment, the model successfully controlled the maximum temperature rise within 10 °C.For cochlear implantation surgery, we also divide the implantation channel into different stages based on the bone density in CT images to identify thermal parameters and calculate drilling strategies. CONCLUSION: This method provides a new strategy for accurate and effective control of borehole heat generation. SIGNIFICANCE: These achievements provide new ideas and directions for research in cochlear implantation surgery and related fields, and are expected to have extensive application in medical practice.


Subject(s)
Cochlear Implantation , Temperature , Cochlear Implantation/methods , Bone and Bones , Hot Temperature
2.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37909334

ABSTRACT

The PI3K/AKT/mTOR pathway is commonly dysregulated in cancer. Rapalogs exhibit modest clinical benefit, likely owing to their lack of effects on 4EBP1. We hypothesized that bi-steric mTORC1-selective inhibitors would have greater potential for clinical benefit than rapalogs in tumors with mTORC1 dysfunction. We assessed this hypothesis in tumor models with high mTORC1 activity both in vitro and in vivo. Bi-steric inhibitors had strong growth inhibition, eliminated phosphorylated 4EBP1, and induced more apoptosis than rapamycin or MLN0128. Multiomics analysis showed extensive effects of the bi-steric inhibitors in comparison with rapamycin. De novo purine synthesis was selectively inhibited by bi-sterics through reduction in JUN and its downstream target PRPS1 and appeared to be the cause of apoptosis. Hence, bi-steric mTORC1-selective inhibitors are a therapeutic strategy to treat tumors driven by mTORC1 hyperactivation.


Subject(s)
MTOR Inhibitors , Phosphatidylinositol 3-Kinases , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Sirolimus/pharmacology , Apoptosis , Cell Proliferation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
3.
Nat Commun ; 14(1): 1214, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36869048

ABSTRACT

Identifying the mechanisms underlying the regulation of immune checkpoint molecules and the therapeutic impact of targeting them in cancer is critical. Here we show that high expression of the immune checkpoint B7-H3 (CD276) and high mTORC1 activity correlate with immunosuppressive phenotypes and worse clinical outcomes in 11,060 TCGA human tumors. We find that mTORC1 upregulates B7-H3 expression via direct phosphorylation of the transcription factor YY2 by p70 S6 kinase. Inhibition of B7-H3 suppresses mTORC1-hyperactive tumor growth via an immune-mediated mechanism involving increased T-cell activity and IFN-γ responses coupled with increased tumor cell expression of MHC-II. CITE-seq reveals strikingly increased cytotoxic CD38+CD39+CD4+ T cells in B7-H3-deficient tumors. In pan-human cancers, a high cytotoxic CD38+CD39+CD4+ T-cell gene signature correlates with better clinical prognosis. These results show that mTORC1-hyperactivity, present in many human tumors including tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), drives B7-H3 expression leading to suppression of cytotoxic CD4+ T cells.


Subject(s)
T-Lymphocytes , Tumor Escape , Humans , Genes, Regulator , Transcription Factors , Mechanistic Target of Rapamycin Complex 1 , B7 Antigens
4.
Beilstein J Org Chem ; 18: 825-836, 2022.
Article in English | MEDLINE | ID: mdl-35923156

ABSTRACT

The changes in absorption and emission of fluorescent materials with the introduction of Lewis acids have been frequently observed due to either physical or chemical interactions. In this mini-review, we elaborate how Lewis acids adjust the optical properties and the bandgap of luminescent materials by simple coordination reactions. It is common that fluorescent materials containing Lewis basic nitrogen heterocycles are more likely to provide the feasible band gap modulation. The essence of such phenomenon originates from Lewis acid-base coordination and adducts, which highly depends on the electron-accepting property of the Lewis acids. This intermolecular mechanism, considered as post-synthesis of new luminescent compounds offers promising applications in sensing and electroluminescence by manipulating the frontier molecular orbital energy levels of organic conjugated materials, simply based on Lewis acid-base chemistry.

5.
Eur Respir J ; 59(4)2022 04.
Article in English | MEDLINE | ID: mdl-34561286

ABSTRACT

BACKGROUND: Biomarkers of disease progression and treatment response are urgently needed for patients with lymphangioleiomyomatosis (LAM). Activity-based nanosensors, an emerging biosensor class, detect dysregulated proteases in vivo and release a reporter to provide a urinary readout of disease. Because proteases are dysregulated in LAM and may directly contribute to lung function decline, activity-based nanosensors may enable quantitative, real-time monitoring of LAM progression and treatment response. We aimed to assess the diagnostic utility of activity-based nanosensors in a pre-clinical model of pulmonary LAM. METHODS: Tsc2-null cells were injected intravenously into female nude mice to establish a mouse model of pulmonary LAM. A library of 14 activity-based nanosensors, designed to detect proteases across multiple catalytic classes, was administered into the lungs of LAM mice and healthy controls, urine was collected, and mass spectrometry was performed to measure nanosensor cleavage products. Mice were then treated with rapamycin and monitored with activity-based nanosensors. Machine learning was performed to distinguish diseased from healthy and treated from untreated mice. RESULTS: Multiple activity-based nanosensors (PP03 (cleaved by metallo, aspartic and cysteine proteases), padjusted<0.0001; PP10 (cleaved by serine, aspartic and cysteine proteases), padjusted=0.017)) were differentially cleaved in diseased and healthy lungs, enabling strong classification with a machine learning model (area under the curve (AUC) 0.95 from healthy). Within 2 days after rapamycin initiation, we observed normalisation of PP03 and PP10 cleavage, and machine learning enabled accurate classification of treatment response (AUC 0.94 from untreated). CONCLUSIONS: Activity-based nanosensors enable noninvasive, real-time monitoring of disease burden and treatment response in a pre-clinical model of LAM.


Subject(s)
Cysteine Proteases , Lymphangioleiomyomatosis , Animals , Cysteine Proteases/therapeutic use , Female , Humans , Lymphangioleiomyomatosis/drug therapy , Mice , Mice, Nude , Peptide Hydrolases/therapeutic use , Sirolimus/therapeutic use
6.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34544857

ABSTRACT

Tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM) are caused by aberrant mechanistic Target of Rapamycin Complex 1 (mTORC1) activation due to loss of either TSC1 or TSC2 Cytokine profiling of TSC2-deficient LAM patient-derived cells revealed striking up-regulation of Interleukin-6 (IL-6). LAM patient plasma contained increased circulating IL-6 compared with healthy controls, and TSC2-deficient cells showed up-regulation of IL-6 transcription and secretion compared to wild-type cells. IL-6 blockade repressed the proliferation and migration of TSC2-deficient cells and reduced oxygen consumption and extracellular acidification. U-13C glucose tracing revealed that IL-6 knockout reduced 3-phosphoserine and serine production in TSC2-deficient cells, implicating IL-6 in de novo serine metabolism. IL-6 knockout reduced expression of phosphoserine aminotransferase 1 (PSAT1), an essential enzyme in serine biosynthesis. Importantly, recombinant IL-6 treatment rescued PSAT1 expression in the TSC2-deficient, IL-6 knockout clones selectively and had no effect on wild-type cells. Treatment with anti-IL-6 (αIL-6) antibody similarly reduced cell proliferation and migration and reduced renal tumors in Tsc2+/- mice while reducing PSAT1 expression. These data reveal a mechanism through which IL-6 regulates serine biosynthesis, with potential relevance to the therapy of tumors with mTORC1 hyperactivity.


Subject(s)
Interleukin-6/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Serine/metabolism , Transaminases/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Animals , Interleukin-6/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Transaminases/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/physiology
7.
Nat Commun ; 12(1): 4245, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253722

ABSTRACT

Tuberous Sclerosis Complex (TSC) is caused by TSC1 or TSC2 mutations, resulting in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). Transcription factor EB (TFEB), a master regulator of lysosome biogenesis, is negatively regulated by mTORC1 through a RAG GTPase-dependent phosphorylation. Here we show that lysosomal biogenesis is increased in TSC-associated renal tumors, pulmonary lymphangioleiomyomatosis, kidneys from Tsc2+/- mice, and TSC1/2-deficient cells via a TFEB-dependent mechanism. Interestingly, in TSC1/2-deficient cells, TFEB is hypo-phosphorylated at mTORC1-dependent sites, indicating that mTORC1 is unable to phosphorylate TFEB in the absence of the TSC1/2 complex. Importantly, overexpression of folliculin (FLCN), a GTPase activating protein for RAGC, increases TFEB phosphorylation at the mTORC1 sites in TSC2-deficient cells. Overexpression of constitutively active RAGC is sufficient to relocalize TFEB to the cytoplasm. These findings establish the TSC proteins as critical regulators of lysosomal biogenesis via TFEB and RAGC and identify TFEB as a driver of the proliferation of TSC2-deficient cells.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Organelle Biogenesis , Tuberous Sclerosis Complex 2 Protein/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Renal Cell/pathology , Cell Nucleus/metabolism , Cell Proliferation , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Kidney Neoplasms/pathology , Lysosomes/ultrastructure , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Tuberous Sclerosis Complex 2 Protein/deficiency , Tumor Suppressor Proteins/metabolism
8.
Oncogene ; 40(1): 112-126, 2021 01.
Article in English | MEDLINE | ID: mdl-33082558

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant tumor suppressor syndrome, characterized by tumor development in multiple organs, including renal angiomyolipoma. Biallelic loss of TSC1 or TSC2 is a known genetic driver of angiomyolipoma development, however, whether an altered transcriptional repertoire contributes to TSC-associated tumorigenesis is unknown. RNA-seq analyses showed that MITF A isoform (MITF-A) was consistently highly expressed in angiomyolipoma, immunohistochemistry showed microphthalmia-associated transcription factor nuclear localization, and Chromatin immuno-Precipitation Sequencing analysis showed that the MITF-A transcriptional start site was highly enriched with H3K27ac marks. Using the angiomyolipoma cell line 621-101, MITF knockout (MITF.KO) and MITF-A overexpressing (MITF.OE) cell lines were generated. MITF.KO cells showed markedly reduced growth and invasion in vitro, and were unable to form xenografted tumors. In contrast, MITF.OE cells grew faster in vitro and as xenografted tumors compared to control cells. RNA-Seq analysis showed that both ID2 and Cysteine-rich angiogenic inducer 61 (CYR61) expression levels were increased in the MITF.OE cells and reduced in the MITF.KO cells, and luciferase assays showed this was due to transcriptional effects. Importantly, CYR61 overexpression rescued MITF.KO cell growth in vitro and tumor growth in vivo. These findings suggest that MITF-A is a transcriptional oncogenic driver of angiomyolipoma tumor development, acting through regulation of CYR61.


Subject(s)
Angiomyolipoma/pathology , Cysteine-Rich Protein 61/genetics , Inhibitor of Differentiation Protein 2/genetics , Kidney Neoplasms/pathology , Microphthalmia-Associated Transcription Factor/genetics , Up-Regulation , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , RNA Isoforms/genetics , Sequence Analysis, RNA , Transcription Initiation Site
10.
Proc Natl Acad Sci U S A ; 117(45): 28056-28067, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33097662

ABSTRACT

The Rac-GEF, P-Rex1, activates Rac1 signaling downstream of G protein-coupled receptors and PI3K. Increased P-Rex1 expression promotes melanoma progression; however, its role in breast cancer is complex, with differing reports of the effect of its expression on disease outcome. To address this we analyzed human databases, undertook gene array expression analysis, and generated unique murine models of P-Rex1 gain or loss of function. Analysis of PREX1 mRNA expression in breast cancer cDNA arrays and a METABRIC cohort revealed that higher PREX1 mRNA in ER+ve/luminal tumors was associated with poor outcome in luminal B cancers. Prex1 deletion in MMTV-neu or MMTV-PyMT mice reduced Rac1 activation in vivo and improved survival. High level MMTV-driven transgenic PREX1 expression resulted in apicobasal polarity defects and increased mammary epithelial cell proliferation associated with hyperplasia and development of de novo mammary tumors. MMTV-PREX1 expression in MMTV-neu mice increased tumor initiation and enhanced metastasis in vivo, but had no effect on primary tumor growth. Pharmacological inhibition of Rac1 or MEK1/2 reduced P-Rex1-driven tumoroid formation and cell invasion. Therefore, P-Rex1 can act as an oncogene and cooperate with HER2/neu to enhance breast cancer initiation and metastasis, despite having no effect on primary tumor growth.


Subject(s)
Guanine Nucleotide Exchange Factors , Mammary Neoplasms, Experimental , Neoplasm Metastasis , Animals , Cell Polarity/genetics , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology
11.
Appl Opt ; 59(4): 1037-1048, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32225240

ABSTRACT

An ultranarrow-bandwidth-optical-receiver-based ultraviolet trifrequency Rayleigh Doppler wind lidar (DWL) technology is proposed that is able to simultaneously detect stratospheric wind with high precision during the daytime. The lidar system is designed, and the principle of wind measurement is analyzed. An ultranarrow-bandwidth element used for suppressing strong background light is designed as an important part of the ultranarrow-bandwidth optical receiver. A three-channel Fabry-Perot interferometer (FPI) is capable of measuring wind speed. A non-polarized beam splitter cube optically contacted on the three-channel FPI can offer a stable splitting ratio. The parameters of the three-channel FPI are optimized. The structure and parameters of the ultranarrow-bandwidth element are designed, and the transmission curve is measured. The transmission curve and stability of the three-channel FPI are validated. The background photon number is collected with the ultranarrow-bandwidth element and with an interference filter (IF) alternately from 08:00 to 18:00. Based on the selected system parameters and measured background photon number, the detection performance of the proposed lidar is simulated. Simulation results show that with 200 m range resolution from 15 to 25 km, 500 m range resolution from 25 to 40 km, and 30 min total accumulation time for paired line-of-sight (LOS) measurement, within $\pm {100}\;{\rm m/s}$±100m/s LOS wind speed range, the daytime LOS wind speed error is below 4.77 m/s from 15 to 40 km altitude. Compared with the traditional IF-based dual-FPI Rayleigh Doppler lidar, the wind speed accuracies are improved by 1.29-16.29 times and the detection altitudes are improved from 23.55 to 40 km with the same wind-detecting precision.

12.
Opt Express ; 27(23): 34230-34246, 2019 Nov 11.
Article in English | MEDLINE | ID: mdl-31878475

ABSTRACT

A well-designed filter assembly is incorporated to an earlier mobile Rayleigh Doppler Lidar developed at University of Science and Technology of China (USTC) for wind measurement round the clock. The filter assembly consists of two cascaded Fabry-Perot Etalons (FPEs) and a narrow-band interference filter (IF), which are optimized to filter out strong solar background radiation during daytime. The high resolution FPE is mainly used to compress the whole bandwidth of the filter assembly, whereas the low resolution FPE with relatively large free spectral range (FSR) is primarily used to block the unwanted periodic transmission peaks of high resolution FPE arising within the narrow-band IF passband. Some test experiments are carried out and demonstrate that the filter assembly have an overall peak transmission of 33.32% with a bandwidth of 2.41 pm at 355 nm. When applying it to the USTC mobile Rayleigh Doppler Lidar, the daytime background is only 3% or less than before. Consequently, the detectable altitude during daytime increases to ∼51 km with wind velocity accuracy of ±7.6 m/s.

13.
Chest ; 156(6): 1062-1067, 2019 12.
Article in English | MEDLINE | ID: mdl-31437431

ABSTRACT

Pulmonary lymphangioleiomyomatosis (LAM) is a rare genetic multisystem disease characterized by the nodular proliferation of smooth muscle-like LAM cells, progressive cystic changes of the lung, lymphatic abnormalities, and renal angiomyolipomas (AMLs). LAM can arise sporadically or in women with the autosomal dominant disorder, tuberous sclerosis complex (TSC), in which hamartomatous tumors of brain, heart, skin, kidney, and lung are found. LAM and TSC are caused by mutations in the TSC1 or TSC2 tumor suppressor genes leading to elevated mechanistic/mammalian target of rapamycin complex activity. Recent data indicate that T cells within LAM nodules and renal AMLs exhibit features of T-cell exhaustion, with coinhibitory receptor programmed cell death protein 1 (PD-1) expression on tumor-infiltrating T cells. Treatment of animal models of TSC and LAM with anti-PD-1 antibodies or with the combination of anti-PD-1 and anti-CTLA4 antibodies has led to remarkable results, suppressing TSC2-null tumor growth and inducing tumor rejection. Here we review our current knowledge about the potential for immunotherapy for the treatment of LAM and TSC and highlight critical unknowns and key next steps.


Subject(s)
Immunotherapy , Lung Neoplasms/therapy , Lymphangioleiomyomatosis/therapy , Tuberous Sclerosis/therapy , Animals , Cell Cycle Checkpoints/drug effects , Forecasting , Humans
14.
Oncogene ; 38(49): 7367-7383, 2019 12.
Article in English | MEDLINE | ID: mdl-31420607

ABSTRACT

miR-29b has been identified as a rapamycin-induced microRNA (miRNA) in Tsc2-deficient, mTORC1-hyperactive cells. The biological significance of this induction of miR-29b is unknown. We have found that miR-29b acts as an oncogenic miRNA in Tsc2-deficient cells: inhibition of miR-29b suppressed cell proliferation, anchorage-independent cell growth, cell migration, invasion, and the growth of Tsc2-deficient tumors in vivo. Importantly, the combination of miR-29b inhibition with rapamycin treatment further inhibited these tumor-associated cellular processes. To gain insight into the molecular mechanisms by which miR-29b promotes tumorigenesis, we used RNA sequencing to identify the tumor suppressor retinoid receptor beta (RARß) as a target gene of miR-29b. We found that miR-29b directly targeted the 3'UTR of RARß. Forced expression of RARß reversed the effects of miR-29b overexpression in proliferation, migration, and invasion, indicating that it is a critical target. miR-29b expression correlated with low RARß expression in renal clear cell carcinomas and bladder urothelial carcinomas, tumors associated with TSC gene mutations. We further identified growth family member 4 (ING4) as a novel interacting partner of RARß. Overexpression of ING4 inhibited the migration and invasion of Tsc2-deficient cells while silencing of ING4 reversed the RARß-mediated suppression of cell migration and invasion. Taken together, our findings reveal a novel miR-29b/RARß/ING4 pathway that regulates tumorigenic properties of Tsc2-deficient cells, and that may serve as a potential therapeutic target for TSC, lymphangioleiomyomatosis (LAM), and other mTORC1-hyperactive tumors.


Subject(s)
Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Expression Regulation , MicroRNAs/genetics , Receptors, Retinoic Acid/metabolism , Sirolimus/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Movement , Cell Proliferation , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Receptors, Retinoic Acid/genetics , Tuberous Sclerosis Complex 2 Protein/physiology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
16.
JCI Insight ; 3(8)2018 04 19.
Article in English | MEDLINE | ID: mdl-29669930

ABSTRACT

Tuberous sclerosis complex (TSC) is an incurable multisystem disease characterized by mTORC1-hyperactive tumors. TSC1/2 mutations also occur in other neoplastic disorders, including lymphangioleiomyomatosis (LAM) and bladder cancer. Whether TSC-associated tumors will respond to immunotherapy is unknown. We report here that the programmed death 1 coinhibitory receptor (PD-1) is upregulated on T cells in renal angiomyolipomas (AML) and pulmonary lymphangioleiomyomatosis (LAM). In C57BL/6J mice injected with syngeneic TSC2-deficient cells, anti-PD-1 alone decreased 105K tumor growth by 67% (P < 0.0001); the combination of PD-1 and CTLA-4 blockade was even more effective in suppressing tumor growth. Anti-PD-1 induced complete rejection of TSC2-deficient 105K tumors in 37% of mice (P < 0.05). Double blockade of PD-1 and CTLA-4 induced rejection in 62% of mice (P < 0.01). TSC2 reexpression in TSC2-deficient TMKOC cells enhanced antitumor immunity by increasing T cell infiltration and production of IFN-γ/TNF-α by T cells, suggesting that TSC2 and mTORC1 play specific roles in the induction of antitumor immunity. Finally, 1 month of anti-PD-1 blockade reduced renal tumor burden by 53% (P < 0.01) in genetically engineered Tsc2+/- mice. Taken together, these data demonstrate for the first time to our knowledge that checkpoint blockade may have clinical efficacy for TSC and LAM, and possibly other benign tumor syndromes, potentially yielding complete and durable clinical responses.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy/methods , Mechanistic Target of Rapamycin Complex 1/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis/genetics , Angiomyolipoma/complications , Angiomyolipoma/genetics , Angiomyolipoma/immunology , Animals , CTLA-4 Antigen/metabolism , Drug Therapy, Combination , Lymphangioleiomyomatosis/complications , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/immunology , Male , Mice , Mice, Inbred C57BL , Mutation , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/etiology , Tuberous Sclerosis/immunology , Tuberous Sclerosis Complex 1 Protein , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/complications , Urinary Bladder Neoplasms/pathology
17.
Hum Mol Genet ; 27(9): 1654-1663, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29509898

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disease caused by germline inactivating mutations of TSC1 or TSC2. In TSC-associated tumors of the brain, heart, skin, kidney and lung, inactivation of both alleles of TSC1 or TSC2 leads to hyperactivation of the mTORC1 pathway. The TSC/mTORC1 pathway is a key regulator of cellular processes related to growth, proliferation and autophagy. We and others have previously found that mTORC1 regulates microRNA biogenesis, but the mechanisms are not fully understood. Microprocessor, a multi-protein complex including the nuclease Drosha, processes the primary miR transcript. Using a dual-luciferase reporter, we found that inhibition of mTORC1 or downregulation of Raptor decreased Microprocessor activity, while loss of TSC2 led to a striking increase (∼5-fold) in Microprocessor activity. To determine the global impact of TSC2 on microRNAs we quantitatively analyzed 752 microRNAs in Tsc2-expressing and Tsc2-deficient cells. Out of 259 microRNAs expressed in both cell lines, 137 were significantly upregulated and 24 were significantly downregulated in Tsc2-deficient cells, consistent with the increased Microprocessor activity. Microprocessor activity is known to be regulated in part by GSK3ß. We found that total GSK3ß levels were higher in Tsc2-deficient cells, and the increase in Microprocessor activity associated with Tsc2 loss was reversed by three different GSK3ß inhibitors. Furthermore, mTOR inhibition increased the levels of phospho-GSK3ß (S9), which negatively affects Microprocessor activity. Taken together these data reveal that TSC2 regulates microRNA biogenesis and Microprocessor activity via GSK3ß.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , MicroRNAs/genetics , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Glycogen Synthase Kinase 3 beta/genetics , HeLa Cells , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , RNA, Small Interfering/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
18.
Oncotarget ; 8(39): 64714-64727, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029388

ABSTRACT

mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro. miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells' clonogenic and anchorage independent growth were reduced by ∼50% (p<0.01) and ∼75% (p<0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% (p<0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo, reducing tumor size by approximately 3-fold (p<0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation (p=0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs.

19.
Cancer Res ; 77(12): 3255-3267, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28512249

ABSTRACT

p62/sequestosome-1 (SQSTM1) is a multifunctional adaptor protein and autophagic substrate that accumulates in cells with hyperactive mTORC1, such as kidney cells with mutations in the tumor suppressor genes tuberous sclerosis complex (TSC)1 or TSC2. Here we report that p62 is a critical mediator of TSC2-driven tumorigenesis, as Tsc2+/- and Tsc2f/f Ksp-CreERT2+ mice crossed to p62-/- mice were protected from renal tumor development. Metabolic profiling revealed that depletion of p62 in Tsc2-null cells decreased intracellular glutamine, glutamate, and glutathione (GSH). p62 positively regulated the glutamine transporter Slc1a5 and increased glutamine uptake in Tsc2-null cells. We also observed p62-dependent changes in Gcl, Gsr, Nqo1, and Srxn1, which were decreased by p62 attenuation and implicated in GSH production and utilization. p62 attenuation altered mitochondrial morphology, reduced mitochondrial membrane polarization and maximal respiration, and increased mitochondrial reactive oxygen species and mitophagy marker PINK1. These mitochondrial phenotypes were rescued by addition of exogenous GSH and overexpression of Sod2, which suppressed indices of mitochondrial damage and promoted growth of Tsc2-null cells. Finally, p62 depletion sensitized Tsc2-null cells to both oxidative stress and direct inhibition of GSH biosynthesis by buthionine sulfoximine. Our findings show how p62 helps maintain intracellular pools of GSH needed to limit mitochondrial dysfunction in tumor cells with elevated mTORC1, highlighting p62 and redox homeostasis as nodal vulnerabilities for therapeutic targeting in these tumors. Cancer Res; 77(12); 3255-67. ©2017 AACR.


Subject(s)
Carcinogenesis/metabolism , Mitochondria/pathology , Multiprotein Complexes/metabolism , Sequestosome-1 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Animals , Carcinogenesis/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Glutathione/biosynthesis , Immunohistochemistry , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
20.
J Biol Chem ; 291(33): 17258-70, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27358402

ABSTRACT

PtdIns(3,4,5)P3-dependent Rac exchanger 1 (PREX1) is a Rac-guanine nucleotide exchange factor (GEF) overexpressed in a significant proportion of human breast cancers that integrates signals from upstream ErbB2/3 and CXCR4 membrane surface receptors. However, the PREX1 domains that facilitate its oncogenic activity and downstream signaling are not completely understood. We identify that ERK1/2 MAPK acts downstream of PREX1 and contributes to PREX1-mediated anchorage-independent cell growth. PREX1 overexpression increased but its shRNA knockdown decreased ERK1/2 phosphorylation in response to EGF/IGF-1 stimulation, resulting in induction of the cell cycle regulators cyclin D1 and p21(WAF1/CIP1) PREX1-mediated ERK1/2 phosphorylation, anchorage-independent cell growth, and cell migration were suppressed by inhibition of MEK1/2/ERK1/2 signaling. PREX1 overexpression reduced staurosporine-induced apoptosis whereas its shRNA knockdown promoted apoptosis in response to staurosporine or the anti-estrogen drug tamoxifen. Expression of wild-type but not GEF-inactive PREX1 increased anchorage-independent cell growth. In addition, mouse xenograft studies revealed that expression of wild-type but not GEF-dead PREX1 resulted in the formation of larger tumors that displayed increased phosphorylation of ERK1/2 but not AKT. The impaired anchorage-independent cell growth, apoptosis, and ERK1/2 signaling observed in stable PREX1 knockdown cells was restored by expression of wild-type but not GEF-dead-PREX1. Therefore, PREX1-Rac-GEF activity is critical for PREX1-dependent anchorage-independent cell growth and xenograft tumor growth and may represent a possible therapeutic target for breast cancers that exhibit PREX1 overexpression.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/biosynthesis , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...