Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 171(7): 1545-1558.e18, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153836

ABSTRACT

mTORC1 is a signal integrator and master regulator of cellular anabolic processes linked to cell growth and survival. Here, we demonstrate that mTORC1 promotes lipid biogenesis via SRPK2, a key regulator of RNA-binding SR proteins. mTORC1-activated S6K1 phosphorylates SRPK2 at Ser494, which primes Ser497 phosphorylation by CK1. These phosphorylation events promote SRPK2 nuclear translocation and phosphorylation of SR proteins. Genome-wide transcriptome analysis reveals that lipid biosynthetic enzymes are among the downstream targets of mTORC1-SRPK2 signaling. Mechanistically, SRPK2 promotes SR protein binding to U1-70K to induce splicing of lipogenic pre-mRNAs. Inhibition of this signaling pathway leads to intron retention of lipogenic genes, which triggers nonsense-mediated mRNA decay. Genetic or pharmacological inhibition of SRPK2 blunts de novo lipid synthesis, thereby suppressing cell growth. These results thus reveal a novel role of mTORC1-SRPK2 signaling in post-transcriptional regulation of lipid metabolism and demonstrate that SRPK2 is a potential therapeutic target for mTORC1-driven metabolic disorders.


Subject(s)
Gene Expression Regulation , Lipogenesis , RNA Processing, Post-Transcriptional , Signal Transduction , Animals , Cell Nucleus/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33536341

ABSTRACT

Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl-/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.


Subject(s)
Carbonic Anhydrase II/genetics , Forkhead Transcription Factors/genetics , Renal Insufficiency/genetics , Tuberous Sclerosis Complex 1 Protein/genetics , Animals , Cysts/genetics , Cysts/pathology , Disease Models, Animal , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mutation/genetics , Proton-Translocating ATPases/genetics , Renal Insufficiency/pathology , TRPP Cation Channels/genetics , Tuberous Sclerosis
3.
BMC Med Educ ; 23(1): 210, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37016345

ABSTRACT

BACKGROUND: Dismantling structural inequities in health care requires that physicians understand the impacts of social determinants of health (SDH). Although many medical schools incorporate SDH education, integration of these principles into the preclinical curriculum remains challenging. METHODS: Students and faculty at the University of Vermont, Larner College of Medicine developed the Social Medicine Theme of the Week (SMTW), a peer-teaching approach to integrating SDH topics across the preclinical curriculum as part of a broader social medicine curriculum. Students created objectives to link SDH-related topics to the weekly curriculum and presented them to the class. Student innovation led to the incorporation of creative online infographics that were published in the curriculum calendar. First year medical students and faculty members were surveyed to assess preferences and educational impact of the SMTW announcements with accompanying infographics. RESULTS: Of the 40 student respondents, 77.5% reported that their knowledge of SDH had improved due to the SMTW. Most students (82.5%) preferred the infographic modality over traditional teaching modalities. Faculty respondents reported limited engagement with the SMTW and, although they supported the need for these objectives, many (61%) found it difficult to integrate SDH content into their class materials. CONCLUSION: Student-led infographics are a popular method of integrating SDH content in the preclinical curriculum that can be optimized through faculty orientation and support. Success for this type of instruction requires opportunities for student developers, integration and formal assessment of objectives, faculty engagement and training, and institutional support for creating and delivering a robust social medicine curriculum.


Subject(s)
Education, Medical, Undergraduate , Students, Medical , Humans , Social Determinants of Health , Curriculum , Faculty , Surveys and Questionnaires
4.
J Med Syst ; 46(7): 48, 2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35670870

ABSTRACT

Justifications for the widespread adoption and integration of an electronic health record (EHR) have long leaned on the purported benefits of the technology. However, the performance of the EHR has been underwhelming relative to the promises of immediate access to relevant patient information, clinical decision supports, computerized ordering, and transferable patient data. In this narrative review, we provide an overview of the historical problems and limitations of the EHR, detail the core principles that define agile processes that may overcome the barriers faced by the current EHR, and re-imagine what an integrated, seamless EHR that serves its users and patients might look like. Moving forward, the EHR should be redesigned using a middle-out framework and empowering dual-type champions to maintain the sustainable diffusion of future innovations.


Subject(s)
Electronic Health Records , Humans
5.
Am J Respir Crit Care Med ; 202(10): 1373-1387, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32603599

ABSTRACT

Rationale: Lymphangioleiomyomatosis (LAM) is a metastatic neoplasm of reproductive-age women associated with mutations in tuberous sclerosis complex genes. LAM causes cystic remodeling of the lung and progressive respiratory failure. The sources and cellular characteristics of LAM cells underlying disease pathogenesis remain elusive.Objectives: Identification and characterization of LAM cells in human lung and uterus using a single-cell approach.Methods: Single-cell and single-nuclei RNA sequencing on LAM (n = 4) and control (n = 7) lungs, immunofluorescence confocal microscopy, ELISA, and aptamer proteomics were used to identify and validate LAMCORE cells and secreted biomarkers, predict cellular origins, and define molecular and cellular networks in LAM.Measurements and Main Results: A unique cell type termed LAMCORE was identified, which was distinct from, but closely related to, lung mesenchymal cells. LAMCORE cells expressing signature genes included known LAM markers such as PMEL, FIGF, CTSK, and MLANA and novel biomarkers validated by aptamer screening, ELISA, and immunofluorescence microscopy. LAM cells in lung and uterus are morphologically indistinguishable and share similar gene expression profiles and biallelic TSC2 mutations, supporting a potential uterine origin for the LAMCORE cell. Effects of LAM on resident pulmonary cell types indicated recruitment and activation of lymphatic endothelial cells.Conclusions: A unique population of LAMCORE cells was identified in lung and uterus of patients with LAM, sharing close transcriptomic identity. LAM cell selective markers, secreted biomarkers, and the predicted cellular molecular features provide new insights into the signaling and transcriptional programs that may serve as diagnostic markers and therapeutic targets to influence the pathogenesis of LAM.


Subject(s)
Biomarkers, Tumor/genetics , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lymphangioleiomyomatosis/diagnosis , Lymphangioleiomyomatosis/genetics , Transcriptome/genetics , Uterine Neoplasms/diagnosis , Uterine Neoplasms/genetics , Adult , Aged , Female , Humans , Middle Aged , Single-Cell Analysis , United States
6.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922083

ABSTRACT

In the connectivity map (CMap) approach to drug repositioning and development, transcriptional signature of disease is constructed by differential gene expression analysis between the diseased tissue or cells and the control. The negative correlation between the transcriptional disease signature and the transcriptional signature of the drug, or a bioactive compound, is assumed to indicate its ability to "reverse" the disease process. A major limitation of traditional CMaP analysis is the use of signatures derived from bulk disease tissues. Since the key driver pathways are most likely dysregulated in only a subset of cells, the "averaged" transcriptional signatures resulting from bulk analysis lack the resolution to effectively identify effective therapeutic agents. The use of single-cell RNA-seq (scRNA-seq) transcriptomic assay facilitates construction of disease signatures that are specific to individual cell types, but methods for using scRNA-seq data in the context of CMaP analysis are lacking. Lymphangioleiomyomatosis (LAM) mutations in TSC1 or TSC2 genes result in the activation of the mTOR complex 1 (mTORC1). The mTORC1 inhibitor Sirolimus is the only FDA-approved drug to treat LAM. Novel therapies for LAM are urgently needed as the disease recurs with discontinuation of the treatment and some patients are insensitive to the drug. We developed methods for constructing disease transcriptional signatures and CMaP analysis using scRNA-seq profiling and applied them in the analysis of scRNA-seq data of lung tissue from naïve and sirolimus-treated LAM patients. New methods successfully implicated mTORC1 inhibitors, including Sirolimus, as capable of reverting the LAM transcriptional signatures. The CMaP analysis mimicking standard bulk-tissue approach failed to detect any connection between the LAM signature and mTORC1 signaling. This indicates that the precise signature derived from scRNA-seq data using our methods is the crucial difference between the success and the failure to identify effective therapeutic treatments in CMaP analysis.


Subject(s)
Biomarkers, Tumor/metabolism , Connectome/methods , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/pathology , Single-Cell Analysis/methods , TOR Serine-Threonine Kinases/metabolism , Antibiotics, Antineoplastic/therapeutic use , Biomarkers, Tumor/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Prognosis , Sequence Analysis, RNA , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/genetics
7.
J Med Syst ; 45(10): 92, 2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34494167

ABSTRACT

The Acute Care Surgery model has been implemented by many hospitals in the United States. As complex adaptive systems, healthcare systems are composed of many interacting elements that respond to intrinsic and extrinsic inputs. Systems level analysis may reveal the underlying organizational structure of tactical block allocations like the Acute Care Surgery model. The purpose of this study is to demonstrate one method to identify a key characteristic of complex adaptive systems in the perioperative services. Start and end times for all surgeries performed at the University of Vermont Medical Center OR1 were extracted for two years prior to the transition to an Acute Care Surgery service and two years following the transition. Histograms were plotted for the inter-event times calculated from the difference between surgical cases. A power law distribution was fit to the post-transition histogram. The Kolmogorov-Smirnov test for goodness-of-fit at 95% level of significance shows the histogram plotted from post-transition inter-event times follows a power law distribution (K-S = 0.088, p = 0.068), indicating a Complex Adaptive System. Our analysis demonstrates that the strategic decision to create an Acute Care Surgery service has direct implications on tactical and operational processes in the perioperative services. Elements of complex adaptive systems can be represented by a power law distributions and similar methods may be applied to identify other processes that operate as complex adaptive systems in perioperative care. To make sustained improvements in the perioperative services, focus on manufacturing-based interventions such as Lean Six Sigma should instead be shifted towards the complex interventions that modify system-specific behaviors described by complex adaptive system principles when power law relationships are present.


Subject(s)
Hospitals , Operating Rooms , Critical Care , Delivery of Health Care , Humans , Total Quality Management , United States
8.
Proc Natl Acad Sci U S A ; 112(1): E21-9, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25524627

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) integrates multiple signals from growth factors, nutrients, and cellular energy status to control a wide range of metabolic processes, including mRNA biogenesis; protein, nucleotide, and lipid synthesis; and autophagy. Deregulation of the mTORC1 pathway is found in cancer as well as genetic disorders such as tuberous sclerosis complex (TSC) and sporadic lymphangioleiomyomatosis. Recent studies have shown that the mTORC1 inhibitor rapamycin and its analogs generally suppress proliferation rather than induce apoptosis. Therefore, it is critical to use alternative strategies to induce death of cells with activated mTORC1. In this study, a small-molecule screen has revealed that the combination of glutaminase (GLS) and heat shock protein 90 (Hsp90) inhibitors selectively triggers death of TSC2-deficient cells. At a mechanistic level, high mTORC1-driven translation rates in TSC1/2-deficient cells, unlike wild-type cells, sensitizes these cells to endoplasmic reticulum (ER) stress. Thus, Hsp90 inhibition drives accumulation of unfolded protein and ER stress. When combining proteotoxic stress with oxidative stress by depletion of the intracellular antioxidant glutathione by GLS inhibition, acute cell death is observed in cells with activated mTORC1 signaling. This study suggests that this combination strategy may have the potential to be developed into a therapeutic use for the treatment of mTORC1-driven tumors.


Subject(s)
Glutaminase/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Animals , Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival/drug effects , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/metabolism , Glutaminase/metabolism , Glutamine/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Oxidation-Reduction/drug effects , Phenotype , Sirolimus/pharmacology , Small Molecule Libraries/pharmacology , Sulfides/pharmacology , Thiadiazoles/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
9.
Am J Respir Cell Mol Biol ; 51(6): 738-49, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24874429

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a female-predominant cystic lung disease that can lead to respiratory failure. LAM cells typically have inactivating tuberous sclerosis complex 2 (TSC2) mutations and mammalian target of rapamycin (mTOR) complex (mTORC) 1 activation. Clinical response to the mTORC1 inhibitors has been limited, prompting a search for additional therapy for LAM. In this study, we investigated the impact of TSC2 on the expression of poly (ADP-ribose) polymerase (PARP)-1 that initiates the DNA repair pathway, and tested the efficacy of PARP1 inhibitors in the survival of TSC2-deficient (TSC2(-)) cells. We analyzed publicly available expression arrays of TSC2(-) cells and validated the findings using real-time RT-PCR, immunoblotting, and immunohistochemistry. We examined the impact of rapamycin and Torin 1 on PARP1 expression. We also tested the effect of PARP1 inhibitors, 8-hydroxy-2-methylquinazoline-4-one and 3,4-dihydro-5[4-(1-piperindinyl)butoxy]-1(2H)-isoquinoline, on the survival of TSC2(-) cells. We identified the up-regulation of PARP1 in TSC2(-) cells relative to cells in which wild-type TSC2 has been reintroduced (TSC2-addback [TSC2(+)] cells). The transcript levels of PARP1 in TSC2(-) cells were not affected by rapamycin. PARP1 levels were increased in TSC2(-) cells, xenograft tumors of rat-derived TSC2(-) cells, renal cystadenomas from Tsc2(+/-) mice, and human LAM nodules. RNA interference of mTOR failed to reduce PARP1 levels. Proliferation and survival of TSC2(-) cells was reduced in response to PARP1 inhibitor treatment, more so than TSC2(+) cells. TSC2(-) cells exhibit higher levels of PARP1 relative to TSC2(+) cells in an mTOR-insensitive manner. PARP1 inhibitors selectively suppress the growth and induce apoptosis of TSC2(-) cells from patients with LAM. Targeting PARP1 may be beneficial in the treatment of LAM and other neoplasm with mTORC1 activation.


Subject(s)
Lung Neoplasms/enzymology , Lymphangioleiomyomatosis/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Quinazolines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line , DNA Repair , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Enzymologic , Humans , Isoquinolines/pharmacology , Lung Neoplasms/drug therapy , Lymphangioleiomyomatosis/drug therapy , Mice, Inbred C57BL , Mice, SCID , Molecular Targeted Therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Rats , Sirolimus/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Up-Regulation , Xenograft Model Antitumor Assays
10.
Monash Bioeth Rev ; 32(1-2): 121-40, 2014.
Article in English | MEDLINE | ID: mdl-25434068

ABSTRACT

In recent years policy makers and public health professionals have described obesity and its associated diseases as a major global public health problem. Bioethicists have tried to address the normative implications of proposed public health interventions by developing guidelines or proposing ethical principles that ethically grounded health policy responses should take into consideration. We are reviewing here relevant literature and conclude that while there are clearly health (and health care cost) implications resulting from the increasing number of seriously obese people across the globe, there appear to be legitimate questions about the scope of the problem as well as questions about whether particular demonstrable correlations are indicative of causations. These empirical questions require further clinical and epidemiological research. We then review currently discussed public health ethics guidance documents and proposals. Suffering from the same conceptual problems that are known features of principle-based bioethics, insofar as their capacity to ground ethically justifiable policies is concerned, they are unsuitable for actual policy development. Even if the empirical questions were resolved, health policy makers could not rely on currently existing prominent public health ethics guidance documents to develop ethically defensible policies. Further empirical and ethics research is necessary to develop ethically defensible public health policies targeting obesity.


Subject(s)
Chronic Disease/prevention & control , Data Collection/ethics , Health Policy , Health Promotion/ethics , Obesity/prevention & control , Public Health/ethics , Bioethical Issues , Body Mass Index , Canada/epidemiology , Chronic Disease/epidemiology , Humans , Obesity/epidemiology , Prevalence
11.
Am J Respir Cell Mol Biol ; 49(1): 135-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23526212

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a destructive lung disease primarily affecting women. Genetic studies indicate that LAM cells carry inactivating tuberous sclerosis complex (TSC)-2 mutations, and metastasize to the lung. We previously discovered that estradiol increases the metastasis of TSC2-deficient cells in mice carrying xenograft tumors. Here, we investigate the molecular basis underlying the estradiol-induced lung metastasis of TSC2-deficient cells, and test the efficacy of Faslodex (an estrogen receptor antagonist) in a preclinical model of LAM. We used a xenograft tumor model in which estradiol induces the lung metastasis of TSC2-deficient cells. We analyzed the impact of Faslodex on tumor size, the extracellular matrix organization, the expression of matrix metalloproteinase (MMP)-2, and lung metastasis. We also examined the effects of estradiol and Faslodex on MMP2 expression and activity in tuberin-deficient cells in vitro. Estradiol resulted in a marked reduction of Type IV collagen deposition in xenograft tumors, associated with 2-fold greater MMP2 concentrations compared with placebo-treated mice. Faslodex normalized the Type IV collagen changes in xenograft tumors, enhanced the survival of the mice, and completely blocked lung metastases. In vitro, estradiol enhanced MMP2 transcripts, protein accumulation, and activity. These estradiol-induced changes in MMP2 were blocked by Faslodex. In TSC2-deficient cells, estradiol increased MMP2 concentrations in vitro and in vivo, and induced extracellular matrix remodeling. Faslodex inhibits the estradiol-induced lung metastasis of TSC2-deficient cells. Targeting estrogen receptors with Faslodex may be of efficacy in the treatment of LAM.


Subject(s)
Estradiol/analogs & derivatives , Estradiol/adverse effects , Extracellular Matrix/drug effects , Lung Neoplasms/secondary , Lymphangioleiomyomatosis/pathology , Airway Remodeling , Animals , Antineoplastic Agents/pharmacology , Collagen Type IV/metabolism , Drug Evaluation, Preclinical , Estradiol/pharmacology , Extracellular Matrix/metabolism , Female , Fulvestrant , Lung/drug effects , Lung/metabolism , Lung/pathology , Lymphangioleiomyomatosis/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, SCID , Rats , Receptors, Estradiol/antagonists & inhibitors , Survival Analysis , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
12.
Sci Adv ; 9(19): eadf8549, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37163604

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare, progressive lung disease that predominantly affects women. LAM cells carry TSC1/TSC2 mutations, causing mTORC1 hyperactivation and uncontrolled cell growth. mTORC1 inhibitors stabilize lung function; however, sustained efficacy requires long-term administration, and some patients fail to tolerate or respond to therapy. Although the genetic basis of LAM is known, mechanisms underlying LAM pathogenesis remain elusive. We integrated single-cell RNA sequencing and single-nuclei ATAC-seq of LAM lungs to construct a gene regulatory network controlling the transcriptional program of LAM cells. We identified activation of uterine-specific HOX-PBX transcriptional programs in pulmonary LAMCORE cells as regulators of cell survival depending upon HOXD11-PBX1 dimerization. Accordingly, blockage of HOXD11-PBX1 dimerization by HXR9 suppressed LAM cell survival in vitro and in vivo. PBX1 regulated STAT1/3, increased the expression of antiapoptotic genes, and promoted LAM cell survival in vitro. The HOX-PBX gene network provides promising targets for treatment of LAM/TSC mTORC1-hyperactive cancers.


Subject(s)
Gene Regulatory Networks , Homeodomain Proteins , Lymphangioleiomyomatosis , Humans , Single-Cell Analysis , Lymphangioleiomyomatosis/metabolism , Lymphangioleiomyomatosis/pathology , Transcription Factors/metabolism , Lung/metabolism , Lung/pathology , Animals , Rats , Neoplasm Metastasis , Multiomics , Female
13.
Nat Commun ; 14(1): 1205, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864068

ABSTRACT

Pulmonary alveolar microlithiasis is an autosomal recessive lung disease caused by a deficiency in the pulmonary epithelial Npt2b sodium-phosphate co-transporter that results in accumulation of phosphate and formation of hydroxyapatite microliths in the alveolar space. The single cell transcriptomic analysis of a pulmonary alveolar microlithiasis lung explant showing a robust osteoclast gene signature in alveolar monocytes and the finding that calcium phosphate microliths contain a rich protein and lipid matrix that includes bone resorbing osteoclast enzymes and other proteins suggested a role for osteoclast-like cells in the host response to microliths. While investigating the mechanisms of microlith clearance, we found that Npt2b modulates pulmonary phosphate homeostasis through effects on alternative phosphate transporter activity and alveolar osteoprotegerin, and that microliths induce osteoclast formation and activation in a receptor activator of nuclear factor-κB ligand and dietary phosphate dependent manner. This work reveals that Npt2b and pulmonary osteoclast-like cells play key roles in pulmonary homeostasis and suggest potential new therapeutic targets for the treatment of lung disease.


Subject(s)
Lung Diseases , Osteogenesis , Humans , Homeostasis , Lung
14.
JCI Insight ; 8(9)2023 05 08.
Article in English | MEDLINE | ID: mdl-36927688

ABSTRACT

Tuberous sclerosis complex (TSC) is characterized by multisystem, low-grade neoplasia involving the lung, kidneys, brain, and heart. Lymphangioleiomyomatosis (LAM) is a progressive pulmonary disease affecting almost exclusively women. TSC and LAM are both caused by mutations in TSC1 and TSC2 that result in mTORC1 hyperactivation. Here, we report that single-cell RNA sequencing of LAM lungs identified activation of genes in the sphingolipid biosynthesis pathway. Accordingly, the expression of acid ceramidase (ASAH1) and dihydroceramide desaturase (DEGS1), key enzymes controlling sphingolipid and ceramide metabolism, was significantly increased in TSC2-null cells. TSC2 negatively regulated the biosynthesis of tumorigenic sphingolipids, and suppression of ASAH1 by shRNA or the inhibitor ARN14976 (17a) resulted in markedly decreased TSC2-null cell viability. In vivo, 17a significantly decreased the growth of TSC2-null cell-derived mouse xenografts and short-term lung colonization by TSC2-null cells. Combined rapamycin and 17a treatment synergistically inhibited renal cystadenoma growth in Tsc2+/- mice, consistent with increased ASAH1 expression and activity being rapamycin insensitive. Collectively, the present study identifies rapamycin-insensitive ASAH1 upregulation in TSC2-null cells and tumors and provides evidence that targeting aberrant sphingolipid biosynthesis pathways has potential therapeutic value in mechanistic target of rapamycin complex 1-hyperactive neoplasms, including TSC and LAM.


Subject(s)
Lung Neoplasms , Tuberous Sclerosis , Humans , Mice , Female , Animals , Tuberous Sclerosis/drug therapy , Tumor Suppressor Proteins/genetics , Up-Regulation , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Acid Ceramidase/therapeutic use , Lung Neoplasms/pathology , Sirolimus/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout
15.
Orphanet J Rare Dis ; 15(1): 209, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807195

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is a genetic disorder that cause tumors to form in many organs. These lesions may lead to epilepsy, autism, developmental delay, renal, and pulmonary failure. Loss of function mutations in TSC1 and TSC2 genes by aberrant activation of the mechanistic target of rapamycin (mTORC1) signaling pathway are the known causes of TSC. Therefore, targeting mTORC1 becomes a most available therapeutic strategy for TSC. Although mTORC1 inhibitor rapamycin and Rapalogs have demonstrated exciting results in the recent clinical trials, however, tumors rebound and upon the discontinuation of the mTORC1 inhibition. Thus, understanding the underlying molecular mechanisms responsible for rapamycin-induced cell survival becomes an urgent need. Identification of additional molecular targets and development more effective remission-inducing therapeutic strategies are necessary for TSC patients. RESULTS: We have discovered an Mitogen-activated protein kinase (MAPK)-evoked positive feedback loop that dampens the efficacy of mTORC1 inhibition. Mechanistically, mTORC1 inhibition increased MEK1-dependent activation of MAPK in TSC-deficient cells. Pharmacological inhibition of MAPK abrogated this feedback loop activation. Importantly, the combinatorial inhibition of mTORC1 and MAPK induces the death of TSC2-deficient cells. CONCLUSIONS: Our results provide a rationale for dual targeting of mTORC1 and MAPK pathways in TSC and other mTORC1 hyperactive neoplasm.


Subject(s)
Tuberous Sclerosis , Cell Survival , Humans , Mitogen-Activated Protein Kinases , Sirolimus/pharmacology , Sirolimus/therapeutic use , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
16.
PLoS One ; 15(2): e0228894, 2020.
Article in English | MEDLINE | ID: mdl-32078667

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a devastating lung disease caused by inactivating gene mutations in either TSC1 or TSC2 that result in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). As LAM occurs predominantly in women during their reproductive age and is exacerbated by pregnancy, the female hormonal environment, and in particular estrogen, is implicated in LAM pathogenesis and progression. However, detailed underlying molecular mechanisms are not well understood. In this study, utilizing human pulmonary LAM specimens and cell culture models of TSC2-deficient LAM patient-derived and rat uterine leiomyoma-derived cells, we tested the hypothesis that estrogen promotes the growth of mTORC1-hyperactive cells through pyruvate kinase M2 (PKM2). Estrogen increased the phosphorylation of PKM2 at Ser37 and induced the nuclear translocation of phospho-PKM2. The estrogen receptor antagonist Faslodex reversed these effects. Restoration of TSC2 inhibited the phosphorylation of PKM2 in an mTORC1 inhibitor-insensitive manner. Finally, accumulation of phosphorylated PKM2 was evident in pulmonary nodule from LAM patients. Together, our data suggest that female predominance of LAM might be at least in part attributed to estrogen stimulation of PKM2-mediated cellular metabolic alterations. Targeting metabolic regulators of PKM2 might have therapeutic benefits for women with LAM and other female-specific neoplasms.


Subject(s)
Estrogens/metabolism , Pyruvate Kinase/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Animals , Cell Line, Tumor , Estrogens/physiology , Female , Humans , Lung/pathology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/physiopathology , Mechanistic Target of Rapamycin Complex 1 , Phosphorylation , Pyruvate Kinase/physiology , Rats , Signal Transduction/drug effects , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Tumor Suppressor Proteins/genetics
17.
Br J Pharmacol ; 176(11): 1680-1699, 2019 06.
Article in English | MEDLINE | ID: mdl-30761522

ABSTRACT

BACKGROUND AND PURPOSE: An up-regulation of COX-2 in malignant gliomas causes excessive synthesis of PGE2 , which is thought to facilitate brain tumour growth and invasion. However, which downstream PGE2 receptor subtype (i.e., EP1 -EP4 ) directly contributes to COX activity-promoted glioma growth remains largely unknown. EXPERIMENTAL APPROACH: Using a publicly available database from The Cancer Genome Atlas research network, we compared the expression of PGE2 signalling-associated genes in human lower grade glioma and glioblastoma multiforme (GBM) samples. The Kaplan-Meier analysis was performed to determine the relationship between their expression and survival probability. A time-resolved FRET method was used to identify the EP subtype that mediates COX-2/PGE2 -initiated cAMP signalling in human GBM cells. Taking advantage of a recently identified novel selective bioavailable brain-permeable small-molecule antagonist, we studied the effect of pharmacological inhibition of the EP2 receptor on glioma cell growth in vitro and in vivo. KEY RESULTS: The EP2 receptor is a key Gαs -coupled receptor that mediates COX-2/PGE2 -initiated cAMP signalling pathways in human malignant glioma cells. Inhibition of EP2 receptors reduced COX-2 activity-driven GBM cell proliferation, invasion, and migration and caused cell cycle arrest at G0-G1 and apoptosis of GBM cells. Glioma cell growth in vivo was also substantially decreased by post-treatment with an EP2 antagonist in both subcutaneous and intracranial tumour models. CONCLUSION AND IMPLICATIONS: Taken together, our results suggest that PGE2 signalling via the EP2 receptor increases the malignant potential of human glioma cells and might represent a novel therapeutic target for GBM.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Glioma/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/metabolism , Glioma/mortality , Humans , Indoles/pharmacology , Kaplan-Meier Estimate , Mice, Inbred C57BL , Mice, Nude , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Signal Transduction/drug effects
18.
Sci Rep ; 9(1): 3015, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816188

ABSTRACT

Tuberous Sclerosis Complex (TSC) and Lymphangioleiomyomatosis (LAM) are caused by inactivating mutations in TSC1 or TSC2, leading to mTORC1 hyperactivation. The mTORC1 inhibitors rapamycin and analogs (rapalogs) are approved for treating of TSC and LAM. Due to their cytostatic and not cytocidal action, discontinuation of treatment leads to tumor regrowth and decline in pulmonary function. Therefore, life-long rapalog treatment is proposed for the control of TSC and LAM lesions, which increases the chances for the development of acquired drug resistance. Understanding the signaling perturbations leading to rapalog resistance is critical for the development of better therapeutic strategies. We developed the first Tsc2-null rapamycin-resistant cell line, ELT3-245, which is highly tumorigenic in mice, and refractory to rapamycin treatment. In vitro ELT3-245 cells exhibit enhanced anchorage-independent cell survival, resistance to anoikis, and loss of epithelial markers. A key alteration in ELT3-245 is increased ß-catenin signaling. We propose that a subset of cells in TSC and LAM lesions have additional signaling aberrations, thus possess the potential to become resistant to rapalogs. Alternatively, when challenged with rapalogs TSC-null cells are reprogrammed to express mesenchymal-like markers. These signaling changes could be further exploited to induce clinically-relevant long-term remissions.


Subject(s)
Drug Resistance/genetics , Mesoderm/metabolism , Tuberous Sclerosis/genetics , Animals , Anoikis/drug effects , Anoikis/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/genetics , Drug Resistance/drug effects , Humans , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mesoderm/drug effects , Mice , Mutation/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology
19.
J Pharm Biomed Anal ; 155: 148-156, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29631075

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a major health threat worldwide. Research focused on molecular events associated with ESCC carcinogenesis for diagnosis, treatment and prevention is needed. Our goal is to discover novel biomarkers and investigate the underlying molecular mechanisms of ESCC progression by employing a global metabolomic approach. Sera from 34 ESCC patients and 32 age and sex matched healthy controls were profiled using two-dimensional liquid chromatography-mass spectrometry (2D LC-MS). We identified 120 differential metabolites in ESCC patient serums compared to healthy controls. Several amino acids, serine, arginine, lysine and histidine were significantly changed in ESCC patients. Most importantly, we found dysregulated lipid metabolism as an important characteristic in ESCC patients. Several free fat acids (FFA) and carnitines were found down-regulated in ESCC patients. Choline was significantly increased and phosphatidylcholines (PC) were significantly decreased in ESCC serum. The high expression of choline and low expression of total PC in patient serum were associated with the high expression of choline kinase (Chok) and activated Kennedy pathway in ESCC cells. Chok expression can serve as a significant biomarker for ESCC prognosis. In conclusion, metabolite profiles in the ESCC patient serum were significantly different from those in the healthy controls. Phosphatidylcholines and Chok, the key enzyme in the PC metabolism pathway, may serve as novel biomarkers for ESCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Choline Kinase/metabolism , Choline/metabolism , Esophageal Neoplasms/metabolism , Amino Acids/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Chromatography, High Pressure Liquid/methods , Disease Progression , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Fatty Acids, Nonesterified/metabolism , Female , Humans , Male , Metabolomics/methods , Middle Aged , Prognosis , Tandem Mass Spectrometry/methods
20.
Oncotarget ; 8(22): 36628-36638, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28410230

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a progressive pulmonary disease that almost exclusively affects women. LAM cells migrate to the lungs, where they cause cystic destruction of lung parenchyma. Mutations in TSC1 or TSC2 lead to the activation of the mammalian target of rapamycin complex-1, a kinase that regulates growth factor-dependent protein translation, cell growth, and metabolism. Insulin-like growth factor binding protein 2 (IGFBP2) binds insulin, IGF1 and IGF2 in circulation, thereby modulating cell survival, migration, and invasion in neoplasms. In this study, we identified that IGFBP2 primarily localized in the nucleus of TSC2-null LAM patient-derived cells in vitro and in vivo. We also showed that nuclear accumulation of IGFBP2 is closely associated with estrogen receptor alpha (ERa) expression. Furthermore, estrogen treatment induced IGFBP2 nuclear translocation in TSC2-null LAM patient-derived cells. Importantly, depletion of IGFBP2 by siRNA reduced cell proliferation, enhanced apoptosis, and decreased migration and invasion of TSC2-null LAM patient-derived cells. More interestingly, depletion of IGFBP2 markedly decreased the phosphorylation of MAPK in LAM patient-derived TSC2-null cells. Collectively, these results suggest that IGFBP2 plays an important role in promoting tumorigenesis, through estrogen and ERalpha signaling pathway. Thus, targeting IGFBP2 may serve as a potential therapeutic strategy for women with LAM and other female gender specific neoplasms.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2/metabolism , Lymphangioleiomyomatosis/metabolism , Lymphangioleiomyomatosis/pathology , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Lymphangioleiomyomatosis/genetics , Phosphorylation , Protein Binding , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL