Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 134(4)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38127441

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a progressive cystic lung disease caused by tuberous sclerosis complex 1/2 (TSC1/2) gene mutations in pulmonary mesenchymal cells, resulting in activation of the mechanistic target of rapamycin complex 1 (mTORC1). A subset of patients with LAM develop pulmonary vascular remodeling and pulmonary hypertension. Little, however, is known regarding how LAM cells communicate with endothelial cells (ECs) to trigger vascular remodeling. In end-stage LAM lung explants, we identified EC dysfunction characterized by increased EC proliferation and migration, defective angiogenesis, and dysmorphic endothelial tube network formation. To model LAM disease, we used an mTORC1 gain-of-function mouse model with a Tsc2 KO (Tsc2KO) specific to lung mesenchyme (Tbx4LME-Cre Tsc2fl/fl), similar to the mesenchyme-specific genetic alterations seen in human disease. As early as 8 weeks of age, ECs from mice exhibited marked transcriptomic changes despite an absence of morphological changes to the distal lung microvasculature. In contrast, 1-year-old Tbx4LME-Cre Tsc2fl/fl mice spontaneously developed pulmonary vascular remodeling with increased medial thickness. Single-cell RNA-Seq of 1-year-old mouse lung cells identified paracrine ligands originating from Tsc2KO mesenchyme, which can signal through receptors in arterial ECs. These ECs had transcriptionally altered genes including those in pathways associated with blood vessel remodeling. The proposed pathophysiologic mesenchymal ligand-EC receptor crosstalk highlights the importance of an altered mesenchymal cell/EC axis in LAM and other hyperactive mTORC1-driven diseases. Since ECs in patients with LAM and in Tbx4LME-Cre Tsc2fl/fl mice did not harbor TSC2 mutations, our study demonstrates that constitutively active mTORC1 lung mesenchymal cells orchestrated dysfunctional EC responses that contributed to pulmonary vascular remodeling.


Subject(s)
Lymphangioleiomyomatosis , Tuberous Sclerosis Complex 2 Protein , Tuberous Sclerosis , Tumor Suppressor Proteins , Animals , Humans , Infant , Mice , Endothelial Cells/metabolism , Lung/metabolism , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesoderm/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vascular Remodeling/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism , In Vitro Techniques
2.
Sci Rep ; 11(1): 8205, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859248

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A-D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cells, Cultured , Humans , Lung/blood supply , Lung/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Vasoconstriction/genetics
3.
Cancer Res ; 81(8): 2086-2100, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33593821

ABSTRACT

Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the "Repurposing" library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2-/- mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2-/- MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2-/- MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2-/- MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. SIGNIFICANCE: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.


Subject(s)
Diacylglycerol Kinase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lymphangioleiomyomatosis/drug therapy , Pinocytosis/drug effects , Ritanserin/pharmacology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis/drug therapy , Angiolipoma/genetics , Animals , Autophagy/drug effects , Cell Proliferation , Chloroquine/pharmacology , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Down-Regulation , Drug Synergism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Kidney Neoplasms/genetics , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/etiology , Lymphangioleiomyomatosis/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Nutrients/metabolism , Phosphatidic Acids/metabolism , Phospholipids/metabolism , Pinocytosis/physiology , Tuberous Sclerosis/complications
4.
Nat Commun ; 11(1): 5640, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159078

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare fatal cystic lung disease due to bi-allelic inactivating mutations in tuberous sclerosis complex (TSC1/TSC2) genes coding for suppressors of the mechanistic target of rapamycin complex 1 (mTORC1). The origin of LAM cells is still unknown. Here, we profile a LAM lung compared to an age- and sex-matched healthy control lung as a hypothesis-generating approach to identify cell subtypes that are specific to LAM. Our single-cell RNA sequencing (scRNA-seq) analysis reveals novel mesenchymal and transitional alveolar epithelial states unique to LAM lung. This analysis identifies a mesenchymal cell hub coordinating the LAM disease phenotype. Mesenchymal-restricted deletion of Tsc2 in the mouse lung produces a mTORC1-driven pulmonary phenotype, with a progressive disruption of alveolar structure, a decline in pulmonary function, increase of rapamycin-sensitive expression of WNT ligands, and profound female-specific changes in mesenchymal and epithelial lung cell gene expression. Genetic inactivation of WNT signaling reverses age-dependent changes of mTORC1-driven lung phenotype, but WNT activation alone in lung mesenchyme is not sufficient for the development of mouse LAM-like phenotype. The alterations in gene expression are driven by distinctive crosstalk between mesenchymal and epithelial subsets of cells observed in mesenchymal Tsc2-deficient lungs. This study identifies sex- and age-specific gene changes in the mTORC1-activated lung mesenchyme and establishes the importance of the WNT signaling pathway in the mTORC1-driven lung phenotype.


Subject(s)
Lung/metabolism , Lymphangioleiomyomatosis/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesoderm/metabolism , Age Factors , Aged , Animals , Female , Humans , Lung/drug effects , Lung/physiopathology , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mesoderm/drug effects , Mice , Sex Factors , Sirolimus/administration & dosage , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism , Wnt Signaling Pathway
5.
Biomolecules ; 10(1)2019 12 24.
Article in English | MEDLINE | ID: mdl-31878201

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare metastatic cystic lung disease due to a mutation in a TSC tumor suppressor, resulting in hyperactive mTOR growth pathways. Sirolimus (rapamycin), an allosteric mTORC1 inhibitor, is a therapeutic option for women with LAM but it only maintains lung volume during treatment and does not provide benefit for all LAM patients. The two major mTORC1 protein synthesis pathways are via S6K/S6 or 4E-BP/eIF4E activation. We aimed to investigate rapamycin in combination with compounds that target associated growth pathways, with the potential to be additive to rapamycin. In this study we demonstrated that rapamycin, at a clinically tolerable concentration (10 nM), inhibited the phosphorylation of S6, but not the critical eIF4E releasing Thr 37/46 phosphorylation sites of 4E-BP1 in TSC2-deficient LAM-derived cells. We also characterized the abundant protein expression of peIF4E within LAM lesions. A selective MNK1/2 inhibitor eFT508 inhibited the phosphorylation of eIF4E but did not reduce TSC2-null cell growth. In contrast, a PI3K/mTOR inhibitor omipalisib blocked the phosphorylation of Akt and both S6K/S6 and 4E-BP/eIF4E branches, and additively decreased the growth of TSC2-null cells with rapamycin. Omipalisib, or another inhibitor of both major mTORC1 growth pathways and pAkt, might provide therapeutic options for TSC2-deficient cancers including, but not limited to, LAM.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis Complex 2 Protein/deficiency , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridazines , Quinolines/pharmacology , Sirolimus/pharmacology , Sulfonamides/pharmacology
6.
Am J Respir Cell Mol Biol ; 59(6): 723-732, 2018 12.
Article in English | MEDLINE | ID: mdl-30095976

ABSTRACT

Pulmonary lymphangioleiomyomatosis (LAM) is a slow-progressing metastatic disease that is driven by mutations in the tumor suppressor tuberous sclerosis complex 1/2 (TSC1/2). Rapamycin inhibits LAM cell proliferation and is the only approved treatment, but it cannot cause the regression of existing lesions and can only stabilize the disease. However, in other cancers, immunotherapies such as checkpoint blockade against PD-1 and its ligand PD-L1 have shown promise in causing tumor regression and even curing some patients. Thus, we asked whether PD-L1 has a role in LAM progression. In vitro, PD-L1 expression in murine Tsc2-null cells is unaffected by mTOR inhibition with torin but can be upregulated by IFN-γ. Using immunohistochemistry and single-cell flow cytometry, we found increased PD-L1 expression both in human lung tissue from patients with LAM and in Tsc2-null lesions in a murine model of LAM. In this model, PD-L1 is highly expressed in the lung by antigen-presenting and stromal cells, and activated T cells expressing PD-1 infiltrate the affected lung. In vivo treatment with anti-PD-1 antibody significantly prolongs mouse survival in the model of LAM. Together, these data demonstrate that PD-1/PD-L1-mediated immunosuppression may occur in LAM, and suggest new opportunities for therapeutic targeting that may provide benefits beyond those of rapamycin.


Subject(s)
B7-H1 Antigen/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , Lymphangioleiomyomatosis/metabolism , Tuberous Sclerosis/metabolism , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/immunology , Case-Control Studies , Cell Proliferation , Disease Models, Animal , Humans , Lung/drug effects , Lung/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/immunology , Lymphangioleiomyomatosis/pathology , Mice , Mice, Inbred C57BL , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/immunology , Tuberous Sclerosis/pathology , Up-Regulation
7.
PLoS One ; 13(5): e0197105, 2018.
Article in English | MEDLINE | ID: mdl-29758070

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare, almost exclusively female lung disease linked to inactivating mutations in tuberous sclerosis complex 2 (TSC2), a tumor suppressor gene that controls cell metabolic state and growth via regulation of the mechanistic target of rapamycin (mTORC1) signaling. mTORC1 is frequently activated in human cancers and, although the mTORC1 inhibitor rapamycin has a cytostatic effect, it is, in general, unable to elicit a robust curative effect or tumor regression. Using RNA-Seq, we identified (1) Insulin-like Growth Factor (IGF2) as one of the genes with the highest fold-change difference between human TSC2-null and TSC2-expressing angiomyolipoma cells from a patient with LAM, and (2) the mouse IGF2 homolog Igf2, as a top-ranking gene according to fold change between Tsc2-/- and Tsc2+/+ mouse embryo fibroblasts (MEFs). We extended transcript-level findings to protein level, observing increased Igf2 protein expression and Igf2 secretion by Tsc2-/- MEFs. Increased Igf2 expression was not due to epigenetic imprinting, but was partially mediated through the Stat3 pathway and was completely insensitive to rapamycin treatment. An siRNA-mediated decrease of Igf2 resulted in decreased Stat3 phosphorylation, suggesting presence of an autocrine Igf2/Stat3 amplification cycle in Tsc2-/- MEFs. In human pulmonary LAM lesions and metastatic cell clusters, high levels of IGF2 were associated with mTORC1 activation. In addition, treatment of three primary IGF2-expressing LAM lung cell lines with rapamycin did not result in IGF2 level changes. Thus, targeting of IGF2 signaling may be of therapeutic value to LAM patients, particularly those who are unresponsive to rapamycin.


Subject(s)
Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/biosynthesis , Lung Neoplasms/metabolism , Signal Transduction , Sirolimus/pharmacology , Tumor Suppressor Proteins/deficiency , Animals , Cell Line, Tumor , Embryo, Mammalian/pathology , Fibroblasts/pathology , Humans , Insulin-Like Growth Factor II/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphangioleiomyomatosis , Mice , Mice, Knockout , Tuberous Sclerosis Complex 2 Protein
8.
Am J Physiol Lung Cell Mol Physiol ; 309(12): L1447-54, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26432869

ABSTRACT

Pulmonary lymphangioleiomyomatosis (LAM), a rare progressive lung disease associated with mutations of the tuberous sclerosis complex 2 (Tsc2) tumor suppressor gene, manifests by neoplastic growth of LAM cells, induction of cystic lung destruction, and respiratory failure. LAM severity correlates with upregulation in serum of the prolymphangiogenic vascular endothelial growth factor D (VEGF-D) that distinguishes LAM from other cystic diseases. The goals of our study was to determine whether Tsc2 deficiency upregulates VEGF-D, and whether axitinib, the Food and Drug Administration-approved small-molecule inhibitor of VEGF receptor (VEGFR) signaling, will reduce Tsc2-null lung lesion growth in a mouse model of LAM. Our data demonstrate upregulation of VEGF-D in the serum and lung lining in mice with Tsc2-null lesions. Progressive growth of Tsc2-null lesions induces recruitment and activation of inflammatory cells and increased nitric oxide production. Recruited cells isolated from the lung lining of mice with Tsc2-null lesions demonstrate upregulated expression of provasculogenic Vegfa, prolymphangiogenic Figf, and proinflammatory Nos2, Il6, and Ccl2 genes. Importantly, axitinib is an effective inhibitor of Tsc2-null lesion growth and inflammatory cell recruitment, which correlates with reduced VEGF-D levels in serum and lung lining. Our data demonstrate that pharmacological inhibition of VEGFR signaling with axitinib inhibits Tsc2-null lesion growth, attenuates recruitment and activation of inflammatory cells, and reduces VEGF-D levels systemically and in the lung lining. Our study suggests a potential therapeutic benefit of inhibition of VEGFR signaling for treatment of LAM.


Subject(s)
Cell Proliferation/drug effects , Imidazoles/pharmacology , Indazoles/pharmacology , Lung/drug effects , Lymphangioleiomyomatosis/drug therapy , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Axitinib , Cell Line, Tumor , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Lung/metabolism , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Nitric Oxide Synthase Type II/metabolism , Tuberous Sclerosis Complex 2 Protein , Up-Regulation/drug effects , Vascular Endothelial Growth Factor D/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...