Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Pharmacol Exp Ther ; 385(1): 35-49, 2023 04.
Article in English | MEDLINE | ID: mdl-36746610

ABSTRACT

Existing vascular endothelial growth factor-oriented antiangiogenic approaches are known for their high potency. However, significant side effects associated with their use drive the need for novel antiangiogenic strategies. The small GTPase RhoA is an established regulator of actin cytoskeletal dynamics. Previous studies have highlighted the impact of endothelial RhoA pathway on angiogenesis. Rho-associate kinase (ROCK), a direct RhoA effector, is potently inhibited by Fasudil, a clinically relevant ROCK inhibitor. Here, we aimed to target the RhoA signaling in endothelial cells by generating Fasudil-encapsulated CD31-targeting liposomes as a potential antiangiogenic therapy. The liposomes presented desirable characteristics, preferential binding to CD31-expressing HEK293T cells and to endothelial cells, inhibited stress fiber formation and cytoskeletal-related morphometric parameters, and inhibited in vitro angiogenic functions. Overall, this work shows that the nanodelivery-mediated endothelial targeting of RhoA signaling can offer a promising strategy for angiogenesis inhibition in vascular-related diseases. SIGNIFICANCE STATEMENT: Systemic administration of antiangiogenic therapeutics induces side effects to non-targeted tissues. This study, among others, has shown the impact of the RhoA signaling in the endothelial cells and their angiogenic functions. Here, to minimize potential toxicity, this study generated CD31-targeting liposomes with encapsulated Fasudil, a clinically relevant Rho kinase inhibitor, and successfully targeted endothelial cells. In this proof-of-principle study, the efficient Fasudil delivery, its impact on the endothelial signaling, morphometric alterations, and angiogenic functions verify the benefits of site-targeted antiangiogenic therapy.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor A , Humans , Endothelial Cells/metabolism , HEK293 Cells , Liposomes , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Int J Mol Sci ; 21(5)2020 Mar 03.
Article in English | MEDLINE | ID: mdl-32138326

ABSTRACT

The tuberous sclerosis complex (Tsc) proteins regulate the conserved mTORC1 growth regulation pathway. We identified that loss of the Tsc2 gene in mouse inner medullary collecting duct (mIMCD) cells induced a greater than two-fold increase in extracellular vesicle (EV) production compared to the same cells having an intact Tsc axis. We optimized EV isolation using a well-established size exclusion chromatography method to produce high purity EVs. Electron microscopy confirmed the purity and spherical shape of EVs. Both tunable resistive pulse sensing (TRPS) and dynamic light scattering (DLS) demonstrated that the isolated EVs possessed a heterogenous size distribution. Approximately 90% of the EVs were in the 100-250 nm size range, while approximately 10% had a size greater than 250 nm. Western blot analysis using proteins isolated from the EVs revealed the cellular proteins Alix and TSG101, the transmembrane proteins CD63, CD81, and CD9, and the primary cilia Hedgehog signaling-related protein Arl13b. Proteomic analysis of EVs identified a significant difference between the Tsc2-intact and Tsc2-deleted cell that correlated well with the increased production. The EVs may be involved in tissue homeostasis and cause disease by overproduction and altered protein content. The EVs released by renal cyst epithelia in TSC complex may serve as a tool to discover the mechanism of TSC cystogenesis and in developing potential therapeutic strategies.


Subject(s)
Extracellular Vesicles/metabolism , Kidney/metabolism , Tuberous Sclerosis/metabolism , Animals , Blotting, Western , Cell Line , Chromatography, Gel , Extracellular Vesicles/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Protein Binding , Proteomics , Tetraspanin 28/metabolism , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism , Tuberous Sclerosis/genetics
3.
J Immunol ; 198(7): 2989-2999, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228558

ABSTRACT

Relatively little is known about factors that initiate immunosuppression in tumors and act at the interface between tumor cells and host cells. In this article, we report novel immunosuppressive properties of the ribosomal protein S19 (RPS19), which is upregulated in human breast and ovarian cancer cells and released from apoptotic tumor cells, whereupon it interacts with the complement C5a receptor 1 expressed on tumor infiltrating myeloid-derived suppressor cells. This interaction promotes tumor growth by facilitating recruitment of these cells to tumors. RPS19 also induces the production of immunosuppressive cytokines, including TGF-ß, by myeloid-derived suppressor cells in tumor-draining lymph nodes, leading to T cell responses skewed toward Th2 phenotypes. RPS19 promotes generation of regulatory T cells while reducing infiltration of CD8+ T cells into tumors. Reducing RPS19 in tumor cells or blocking the C5a receptor 1-RPS19 interaction decreases RPS19-mediated immunosuppression, impairs tumor growth, and delays the development of tumors in a transgenic model of breast cancer. This work provides initial preclinical evidence for targeting RPS19 for anticancer therapy enhancing antitumor T cell responses.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Receptor, Anaphylatoxin C5a/immunology , Ribosomal Proteins/immunology , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Immunoprecipitation , Mice , T-Lymphocytes/immunology
4.
Cytometry A ; 87(5): 451-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25728685

ABSTRACT

Autophagy dysregulation has been implicated in numerous diseases and many therapeutic agents are known to modulate this pathway. Therefore, the ability to accurately monitor autophagy is critical to understanding its role in the pathogenesis and treatment of many diseases. Recently an imaging flow cytometry method measuring colocalization of microtubule associated protein 1B light chain 3 (LC3) and lysosomal signals via Bright Detail Similarity (BDS) was proposed which enabled evaluation of autophagic processing. However, since BDS only evaluates colocalization of LC3 and lysosomal signals, the number of autophagy organelles was not taken into account. We found that in cells classified as having Low BDS, there was a large degree of variability in accumulation of autophagosomes. Therefore, we developed a new approach wherein BDS was combined with number of LC3+ puncta, which enabled us to distinguish between cells having very few autophagy organelles versus cells with accumulation of autophagosomes or autolysosomes. Using this method, we were able to distinguish and quantify autophagosomes and autolysosomes in breast cancer cells cultured under basal conditions, with inhibition of autophagy using chloroquine, and with induction of autophagy using amino acid starvation. This technique yields additional insight into autophagy processing making it a useful supplement to current techniques.


Subject(s)
Autophagy , Cell Tracking , Flow Cytometry/methods , Cell Line, Tumor , Chloroquine/pharmacology , Green Fluorescent Proteins/chemistry , Humans , Lysosomes/pathology , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Phagosomes/pathology
5.
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
6.
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
7.
Proc Natl Acad Sci U S A ; 106(8): 2635-40, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19202070

ABSTRACT

Lymphangioleiomyomatosis (LAM) is an often fatal disease primarily affecting young women in which tuberin (TSC2)-null cells metastasize to the lungs. The mechanisms underlying the striking female predominance of LAM are unknown. We report here that 17-beta-estradiol (E(2)) causes a 3- to 5-fold increase in pulmonary metastases in male and female mice, respectively, and a striking increase in circulating tumor cells in mice bearing tuberin-null xenograft tumors. E(2)-induced metastasis is associated with activation of p42/44 MAPK and is completely inhibited by treatment with the MEK1/2 inhibitor, CI-1040. In vitro, E(2) inhibits anoikis of tuberin-null cells. Finally, using a bioluminescence approach, we found that E(2) enhances the survival and lung colonization of intravenously injected tuberin-null cells by 3-fold, which is blocked by treatment with CI-1040. Taken together these results reveal a new model for LAM pathogenesis in which activation of MEK-dependent pathways by E(2) leads to pulmonary metastasis via enhanced survival of detached tuberin-null cells.


Subject(s)
Cell Survival/physiology , Estrogens/physiology , Lung Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Animals , Anoikis/physiology , Benzamides/pharmacology , Carrier Proteins/antagonists & inhibitors , Female , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Metastasis , Ovariectomy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Rats , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
8.
Genes Dis ; 9(1): 187-200, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35005118

ABSTRACT

TSC renal cystic disease is poorly understood and has no approved treatment. In a new principal cell-targeted murine model of Tsc cystic disease, the renal cystic epithelium is mostly composed of type A intercalated cells with an intact Tsc2 gene confirmed by sequencing, although these cells exhibit a Tsc-mutant disease phenotype. We used a newly derived targeted murine model in lineage tracing and extracellular vesicle (EV) characterization experiments and a cell culture model in EV characterization and cellular induction experiments to understand TSC cystogenesis. Using lineage tracing experiments, we found principal cells undergo clonal expansion but contribute very few cells to the cyst. We determined that cystic kidneys contain more interstitial EVs than noncystic kidneys, excrete fewer EVs in urine, and contain EVs in cyst fluid. Moreover, the loss of Tsc2 gene in EV-producing cells greatly changes the effect of EVs on renal tubular epithelium, such that the epithelium develops increased secretory and proliferative pathway activity. We demonstate that the mTORC1 pathway activity is independent form the EV production, and that the EV effects for a single cell line can vary significantly. TSC cystogenesis involves significant contribution from genetically intact cells conscripted to the mutant phenotype by mutant cell derived EVs.

9.
Genes Chromosomes Cancer ; 49(3): 282-97, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20013896

ABSTRACT

Specificity protein 1 (SP1) is an essential transcription factor implicated in the regulation of genes that control multiple cellular processes, including cell cycle, apoptosis, and DNA damage. Very few nontranscriptional roles for SP1 have been reported thus far. Using confocal microscopy and centrosome fractionation, we identified SP1 as a centrosomal protein. Sp1-deficient mouse embryonic fibroblasts and cells depleted of SP1 by RNAi have increased centrosome number associated with centriole splitting, decreased microtubule nucleation, chromosome misalignment, formation of multipolar mitotic spindles and micronuclei, and increased incidence of aneuploidy. Using mass spectrometry, we identified P70S6K, an effector of the mTOR/raptor (mTORC1) kinase complex, as a novel interacting protein of SP1. We found that SP1-deficient cells have increased phosphorylation of the P70S6K effector ribosomal protein S6, suggesting that SP1 participates in the regulation of the mTORC1/P70S6K/S6 signaling pathway. We previously reported that aberrant mTORC1 activation leads to supernumerary centrosomes, a phenotype rescued by the mTORC1 inhibitor rapamycin. Similarly, treatment with rapamycin rescued the multiple centrosome phenotype of SP1-deficient cells. Taken together, these data strongly support the hypothesis that SP1 is involved in the control of centrosome number via regulation of the mTORC1 pathway, and predict that loss of SP1 function can lead to aberrant centriole splitting, deregulated mTORC1 signaling, and aneuploidy, thereby contributing to malignant transformation.


Subject(s)
Centrioles/physiology , Chromosomal Instability , Gene Silencing , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sp1 Transcription Factor/metabolism , 3T3 Cells , Animals , Apoptosis , Cell Cycle , Cell Line , Centrosome/physiology , Centrosome/ultrastructure , DNA Damage , Gene Expression Regulation , HeLa Cells , Humans , Mice , RNA Interference/physiology , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , TOR Serine-Threonine Kinases
10.
Front Physiol ; 12: 630933, 2021.
Article in English | MEDLINE | ID: mdl-34262466

ABSTRACT

In tuberous sclerosis complex (TSC), Tsc2 mutations are associated with more severe disease manifestations than Tsc1 mutations and the role of extracellular vesicles (EVs) in this context is not yet studied. We report a comparative analysis of EVs derived from isogenic renal cells except for Tsc1 or Tsc2 gene status and hypothesized that in spite of having similar physical characteristics, EVs modulate signaling pathways differently, thus leading to TSC heterogenicity. We used mouse inner medullary collecting duct (mIMCD3) cells with the Tsc1 (T1G cells) or Tsc2 (T2J cells) gene disrupted by CRISPR/CAS9. EVs were isolated from the cell culture media by size-exclusion column chromatography followed by detailed physical and chemical characterization. Physical characterization of EVs was accessed by tunable resistive pulse sensing and dynamic light scattering, revealing similar average sizes and zeta potentials (at pH 7.4) for EVs from mIMCD3 (123.5 ± 5.7 nm and -16.3 ± 2.1 mV), T1G cells (131.5 ± 8.3 nm and -19.8 ± 2.7 mV), and T2J cells (127.3 ± 4.9 nm and -20.2 ± 2.1 mV). EVs derived from parental mIMCD3 cells and both mutated cell lines were heterogeneous (>90% of EVs < 150 nm) in nature. Immunoblotting detected cilial Hedgehog signaling protein Arl13b; intercellular proteins TSG101 and Alix; and transmembrane proteins CD63, CD9, and CD81. Compared to Tsc2 deletion, Tsc1 deletion cells had reduced EV production and release rates. EVs from Tsc1 mutant cells altered mTORC1, autophagy, and ß-catenin pathways differently than EVs from Tsc2-mutated cells. Quantitative PCR analysis revealed the down regulation of miR-212a-3p and miR-99a-5p in EVs from Tsc2-mutated cells compared to EVs from Tsc1-mutant cells. Thus, EV-derived miR-212-3p and mIR-99a-5p axes may represent therapeutic targets or biomarkers for TSC disease.

11.
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
12.
Mol Cancer Res ; 17(8): 1639-1651, 2019 08.
Article in English | MEDLINE | ID: mdl-31088907

ABSTRACT

We discovered that 90.3% of patients with angiomyolipomas, lymphangioleiomyomatosis (LAM), and tuberous sclerosis complex (TSC) carry the arginine variant of codon 72 (R72) of TP53 and that R72 increases the risk for angiomyolipoma. R72 transactivates NOTCH1 and NODAL better than the proline variant of codon 72 (P72); therefore, the expression of NOTCH1 and NODAL is increased in angiomyolipoma cells that carry R72. The loss of Tp53 and Tsc1 within nestin-expressing cells in mice resulted in the development of renal cell carcinomas (RCC) with high Notch1 and Nodal expression, suggesting that similar downstream mechanisms contribute to tumorigenesis as a result of p53 loss in mice and p53 polymorphism in humans. The loss of murine Tp53 or expression of human R72 contributes to tumorigenesis via enhancing epithelial-to-mesenchymal transition and motility of tumor cells through the Notch and Nodal pathways. IMPLICATIONS: This work revealed unexpected contributions of the p53 polymorphism to the pathogenesis of TSC and established signaling alterations caused by this polymorphism as a target for therapy. We found that the codon 72 TP53 polymorphism contributes to TSC-associated tumorigenesis via Notch and Nodal signaling.


Subject(s)
Carcinogenesis/pathology , Nodal Protein/metabolism , Polymorphism, Single Nucleotide , Receptor, Notch1/metabolism , Tuberous Sclerosis/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Angiomyolipoma/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mutation , Nodal Protein/genetics , Receptor, Notch1/genetics , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
13.
Physiol Rep ; 7(2): e13983, 2019 01.
Article in English | MEDLINE | ID: mdl-30675765

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor predisposition syndrome with significant renal cystic and solid tumor disease. While the most common renal tumor in TSC, the angiomyolipoma, exhibits a loss of heterozygosity associated with disease, we have discovered that the renal cystic epithelium is composed of type A intercalated cells that have an intact Tsc gene that have been induced to exhibit Tsc-mutant disease phenotype. This mechanism appears to be different than that for ADPKD. The murine models described here closely resemble the human disease and both appear to be mTORC1 inhibitor responsive. The induction signaling driving cystogenesis may be mediated by extracellular vesicle trafficking.


Subject(s)
Kidney Diseases, Cystic/pathology , Tuberous Sclerosis/pathology , Animals , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Female , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Tuberous Sclerosis/genetics , Tuberous Sclerosis/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
14.
Sci Rep ; 7(1): 16697, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196670

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare autosomal dominant disorder causing benign tumors in the brain and other vital organs. The genes implicated in disease development are TSC1 and TSC2. Here, we have performed mutational analysis followed by a genotype-phenotype correlation study based on the clinical characteristics of the affected individuals. Twenty unrelated probands or families from Greece have been analyzed, of whom 13 had definite TSC, whereas another 7 had a possible TSC diagnosis. Using direct sequencing, we have identified pathogenic mutations in 13 patients/families (6 in TSC1 and 7 in TSC2), 5 of which were novel. The mutation identification rate for patients with definite TSC was 85%, but only 29% for the ones with a possible TSC diagnosis. Multiplex ligation-dependent probe amplification (MLPA) did not reveal any genomic rearrangements in TSC1 and TSC2 in the samples with no mutations identified. In general, TSC2 disease was more severe than TSC1, with more subependymal giant cell astrocytomas and angiomyolipomas, higher incidence of pharmacoresistant epileptic seizures, and more severe neuropsychiatric disorders. To our knowledge, this is the first comprehensive TSC1 and TSC2 mutational analysis carried out in TSC patients in Greece.


Subject(s)
Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis/pathology , Adult , Child , DNA Mutational Analysis , Exons , Female , Gene Deletion , Genetic Association Studies , Greece , Humans , Male , Mutation, Missense , Pedigree , Protein Structure, Tertiary , Tuberous Sclerosis/genetics
15.
Oncogene ; 24(50): 7475-81, 2005 Nov 14.
Article in English | MEDLINE | ID: mdl-16288294

ABSTRACT

The most exciting advances in the tuberous sclerosis complex (TSC) field occurred in 1993 and 1997 with the cloning of the TSC2 and TSC1 genes, respectively, and in 2003 with the identification of Rheb as the target of tuberin's (TSC2) GTPase activating protein (GAP) domain. Rheb has a dual role: it activates mTOR and inactivates B-Raf. Activation of mTOR leads to increased protein synthesis through phosphorylation of p70S6K and 4E-BP1. Upon insulin or growth factor stimulation, tuberin is phosphorylated by several kinases, including AKT/PKB, thereby suppressing its GAP activity and activating mTOR. Phosphorylation of hamartin (TSC1) by CDK1 also negatively regulates the activity of the hamartin/tuberin complex. Despite these biochemical advances, exactly how mutations in TSC1 or TSC2 lead to the clinical manifestations of TSC is far from being understood. Two of the most unusual phenotypes in TSC are the apparent metastasis of benign cells carrying TSC1 and TSC2 mutations, resulting in pulmonary lymphangiomyomatosis, and the ability of cells with TSC1 or TSC2 mutations to differentiate into the separate components of renal angiomyolipomas (vessels, smooth muscle and fat). We will discuss how the TSC signaling pathways are affected by mutations in TSC1 or TSC2, focusing on how these mutations may lead to the renal and pulmonary manifestations of TSC.


Subject(s)
Lymphangioleiomyomatosis/genetics , Signal Transduction , Tuberous Sclerosis/genetics , Tuberous Sclerosis/physiopathology , Tumor Suppressor Proteins/genetics , Cell Proliferation , Humans , Lymphangioleiomyomatosis/physiopathology , Mutation , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/physiology
16.
Methods Mol Med ; 126: 185-96, 2006.
Article in English | MEDLINE | ID: mdl-16930013

ABSTRACT

Tuberous sclerosis complex (TSC) is a neurocutaneous syndrome characterized by seizures, mental retardation, and benign tumors of many organs, including the brain, kidneys, skin, retina, and heart. TSC is caused by mutations in the TSC1 and TSC2 tumor suppressor genes. The genes follow the two-hit model for tumorigenesis, with germline mutations inactivating one allele and somatic mutations inactivating the remaining wild-type allele. Allelic loss (also called loss of heterozygosity [LOH]) in the 9q34 and 16p13 regions has been found in many tumor types from TSC patients. Cardiac rhabdomyomas are frequently found in infants with TSC. Because rhabdomyomas often spontaneously regress, access to fresh tissue is limited. In this chapter, we present methodology for detection of genetic inactivation of TSC1 and TSC2 in paraffin-embedded archival tissues. The template DNA is obtained either by direct scraping of tissue or after laser capture microdissection. LOH analysis is performed after polymerase chain reaction amplification of microsatellite markers in the 9q34 and 16p13 regions and denaturing polyacrylamide gel electrophoresis. Mutation detection is performed using single-strand conformation polymorphisms on mutation detection enhancement gels. Finally, variant bands are amplified and analyzed by direct sequencing.


Subject(s)
DNA Mutational Analysis/methods , Genes, Tumor Suppressor , Mutation/genetics , Tissue Banks , DNA/genetics , DNA/isolation & purification , Humans , Infant , Loss of Heterozygosity/genetics , Microsatellite Repeats/genetics , Paraffin Embedding , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
17.
Oncogene ; 21(55): 8470-6, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12466966

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor suppressor gene syndrome characterized by seizures, mental retardation, autism, and tumors of the brain, kidney, heart, retina, and skin. TSC is caused by mutations in either TSC1 or TSC2, both of which are tumor suppressor genes. Hamartin, the protein product of TSC1, was found to interact with the ezrin-radixin-moesin family of cytoskeletal proteins and to activate the small GTPase Rho. To determine whether tuberin, the TSC2 product, can also activate Rho, we stably expressed full-length human tuberin in two cell types: MDCK cells and ELT3 cells. ELT3 cells lack endogenous tuberin expression. We found that expression of human tuberin in both MDCK and ELT3 cells was associated with an increase in the amount of Rho-GTP, but not in Rac1-GTP or cdc42-GTP. Tuberin expression increased cell adhesion in both cell types, and decreased chemotactic cell migration in ELT3 cells. In MDCK cells, there was a decrease in the amount of total Focal Adhesion Kinase (FAK) and an increase in the fraction of phosphorylated FAK. These findings demonstrate for the first time that tuberin activates Rho and regulates cell adhesion and migration. Pathways involving Rho activation may have relevance to the clinical manifestations of TSC, including pulmonary lymphangioleiomyomatosis.


Subject(s)
Cell Adhesion/genetics , Genes, Tumor Suppressor , Repressor Proteins/genetics , rho GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Movement/genetics , Dogs , Gene Expression Regulation, Neoplastic , Humans , Proteins/genetics , Rats , Transfection , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
18.
Cell Cycle ; 14(3): 399-407, 2015.
Article in English | MEDLINE | ID: mdl-25565629

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) increases translation, cell size and angiogenesis, and inhibits autophagy. mTORC1 is negatively regulated by hamartin and tuberin, the protein products of the tumor suppressors TSC1 and TSC2 that are mutated in Tuberous Sclerosis Complex (TSC) and sporadic Lymphangioleiomyomatosis (LAM). Hamartin interacts with the centrosomal and mitotic kinase polo-like kinase 1 (PLK1). Hamartin and tuberin deficient cells have abnormalities in centrosome duplication, mitotic progression, and cytokinesis, suggesting that the hamartin/tuberin heterodimer and mTORC1 signaling are involved in centrosome biology and mitosis. Here we report that PLK1 protein levels are increased in hamartin and tuberin deficient cells and LAM patient-derived specimens, and that this increase is rapamycin-sensitive. Pharmacological inhibition of PLK1 by the small-molecule inhibitor BI-2536 significantly decreased the viability and clonogenic survival of hamartin and tuberin deficient cells, which was associated with increased apoptosis. BI-2536 increased p62, LC3B-I and GFP-LC3 punctae, and inhibited HBSS-induced degradation of p62, suggesting that PLK1 inhibition attenuates autophagy. Finally, PLK1 inhibition repressed the expression and protein levels of key autophagy genes and proteins and the protein levels of Bcl(-)2 family members, suggesting that PLK1 regulates both autophagic and apoptotic responses. Taken together, our data point toward a previously unrecognized role of PLK1 on the survival of cells with mTORC1 hyperactivation, and the potential use of PLK1 inhibitors as novel therapeutics for tumors with dysregulated mTORC1 signaling, including TSC and LAM.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis/genetics , Autophagy/genetics , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Clone Cells , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Sirolimus/pharmacology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Polo-Like Kinase 1
19.
J Child Neurol ; 19(9): 710-5, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15563018

ABSTRACT

This review is focused on pathways and mechanisms that might provide molecular links between the pathogenesis of renal and pulmonary disease in tuberous sclerosis complex and the pathogenesis of the neurologic manifestations of tuberous sclerosis complex. Tuberous sclerosis complex is an autosomal dominant disorder in which the manifestations can include seizures; mental retardation; autism; benign tumors of the brain, retina, skin, and kidneys; and pulmonary lymphangiomyomatosis. Lymphangiomyomatosis is a life-threatening lung disease affecting almost exclusively young women. Genetic data have demonstrated that the cells giving rise to renal angiomyolipomas, the most frequent tumor type in patients with tuberous sclerosis complex, exhibit differentiation plasticity. Genetic studies have also shown that the benign smooth muscle cells of angiomyolipomas and pulmonary lymphangiomyomatosis have the ability to migrate or metastasize to other organs. These findings indicate that hamartin and tuberin play functional roles in the regulation of cell migration and differentiation. The biochemical pathways responsible for these effects are not yet fully understood but might involve dysregulation of the small guanosine triphosphatase Rho. Similar pathways might contribute to aberrant neuronal differentiation and migration in tuberous sclerosis complex.


Subject(s)
Cell Differentiation , Cell Movement , Kidney Diseases/physiopathology , Tuberous Sclerosis/physiopathology , Child , Humans , Lymphangioleiomyomatosis/physiopathology , Muscle, Smooth/cytology , Proteins/pharmacology , Repressor Proteins/pharmacology , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
20.
Cell Cycle ; 9(6): 1143-55, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20237422

ABSTRACT

Studies of the role of tuberous sclerosis complex (TSC) proteins (TSC1/TSC2) in pathology have focused mainly on their capacity to regulate translation and cell growth, but their relationship with alterations of cellular structures and the cell cycle is not yet fully understood. The transforming acidic coiled-coil (TACC) domain-containing proteins are central players in structures and processes connected to the centrosome. Here, TACC3 interactome mapping identified TSC2 and 15 other physical interactors, including the evolutionary conserved interactions with ch-TOG/CKAP5 and FAM161B. TACC3 and TSC2 co-localize and co-purify with components of the nuclear envelope, and their deficiency causes morphological alterations of this structure. During cell division, TACC3 is necessary for the proper localization of phospho-Ser939 TSC2 at spindle poles and cytokinetic bridges. Accordingly, abscission alterations and increased frequency of binucleated cells were observed in Tacc3- and Tsc2-deficient cells relative to controls. In regulating cell division, TSC2 acts epistatically to TACC3 and, in addition to canonical TSC/mTOR signaling and cytokinetic associations, converges to the early mitotic checkpoint mediated by CHFR, consistently with nuclear envelope associations. Our findings link TACC3 to novel structural and cell division functions of TSC2, which may provide additional explanations for the clinical and pathological manifestations of lymphangioleiomyomatosis (LAM) disease and TSC syndrome, including the greater clinical severity of TSC2 mutations compared to TSC1 mutations.


Subject(s)
Cell Division , Microtubule-Associated Proteins/metabolism , Nuclear Envelope/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Nucleus/metabolism , Cell Survival , Centrosome/metabolism , Cytokinesis , Epistasis, Genetic , HEK293 Cells , HeLa Cells , Humans , Mice , Microtubule-Associated Proteins/deficiency , Mitosis , Phosphoserine/metabolism , Protein Binding , Protein Transport , Spindle Apparatus/metabolism , Subcellular Fractions/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL