Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
EMBO J ; 37(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29438978

ABSTRACT

TDP-43 is an RNA-binding protein active in splicing that concentrates into membraneless ribonucleoprotein granules and forms aggregates in amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. Although best known for its predominantly disordered C-terminal domain which mediates ALS inclusions, TDP-43 has a globular N-terminal domain (NTD). Here, we show that TDP-43 NTD assembles into head-to-tail linear chains and that phosphomimetic substitution at S48 disrupts TDP-43 polymeric assembly, discourages liquid-liquid phase separation (LLPS) in vitro, fluidizes liquid-liquid phase separated nuclear TDP-43 reporter constructs in cells, and disrupts RNA splicing activity. Finally, we present the solution NMR structure of a head-to-tail NTD dimer comprised of two engineered variants that allow saturation of the native polymerization interface while disrupting higher-order polymerization. These data provide structural detail for the established mechanistic role of the well-folded TDP-43 NTD in splicing and link this function to LLPS. In addition, the fusion-tag solubilized, recombinant form of TDP-43 full-length protein developed here will enable future phase separation and in vitro biochemical assays on TDP-43 function and interactions that have been hampered in the past by TDP-43 aggregation.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Aggregation, Pathological/genetics , Protein Domains/genetics , RNA Splicing/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Polymerization , Polymers/metabolism , Protein Aggregation, Pathological/pathology
2.
J Biol Chem ; 294(17): 6696-6709, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30824544

ABSTRACT

Aggregates of the RNA-binding protein TDP-43 (TAR DNA-binding protein) are a hallmark of the overlapping neurodegenerative disorders amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. The process of TDP-43 aggregation remains poorly understood, and whether it includes formation of intermediate complexes is unknown. Here, we analyzed aggregates derived from purified TDP-43 under semidenaturing conditions, identifying distinct oligomeric complexes at the initial time points before the formation of large aggregates. We found that this early oligomerization stage is primarily driven by TDP-43's RNA-binding region. Specific binding to GU-rich RNA strongly inhibited both TDP-43 oligomerization and aggregation, suggesting that RNA interactions are critical for maintaining TDP-43 solubility. Moreover, we analyzed TDP-43 liquid-liquid phase separation and detected similar detergent-resistant oligomers upon maturation of liquid droplets into solid-like fibrils. These results strongly suggest that the oligomers form during the early steps of TDP-43 misfolding. Importantly, the ALS-linked TDP-43 mutations A315T and M337V significantly accelerate aggregation, rapidly decreasing the monomeric population and shortening the oligomeric phase. We also show that aggregates generated from purified TDP-43 seed intracellular aggregation detected by established TDP-43 pathology markers. Remarkably, cytoplasmic aggregate seeding was detected earlier for the A315T and M337V variants and was 50% more widespread than for WT TDP-43 aggregates. We provide evidence for an initial step of TDP-43 self-assembly into intermediate oligomeric complexes, whereby these complexes may provide a scaffold for aggregation. This process is altered by ALS-linked mutations, underscoring the role of perturbations in TDP-43 homeostasis in protein aggregation and ALS-FTD pathogenesis.


Subject(s)
Biopolymers/metabolism , DNA-Binding Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Biomarkers/metabolism , DNA-Binding Proteins/genetics , Disulfides/metabolism , HEK293 Cells , Humans , Molecular Weight , Mutation , Phase Transition , Protein Folding , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism
3.
J Biol Chem ; 292(12): 5089-5100, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28167528

ABSTRACT

TAR DNA-binding protein (TDP-43) is a highly conserved and essential DNA- and RNA-binding protein that controls gene expression through RNA processing, in particular, regulation of splicing. Intracellular aggregation of TDP-43 is a hallmark of amyotrophic lateral sclerosis and ubiquitin-positive frontotemporal lobar degeneration. This TDP-43 pathology is also present in other types of neurodegeneration including Alzheimer's disease. We report here that TDP-43 is a substrate of MEK, a central kinase in the MAPK/ERK signaling pathway. TDP-43 dual phosphorylation by MEK, at threonine 153 and tyrosine 155 (p-T153/Y155), was dramatically increased by the heat shock response (HSR) in human cells. HSR promotes cell survival under proteotoxic conditions by maintaining protein homeostasis and preventing protein misfolding. MEK is activated by HSR and contributes to the regulation of proteome stability. Phosphorylated TDP-43 was not associated with TDP-43 aggregation, and p-T153/Y155 remained soluble under conditions that promote protein misfolding. We found that active MEK significantly alters TDP-43-regulated splicing and that phosphomimetic substitutions at these two residues reduce binding to GU-rich RNA. Cellular imaging using a phospho-specific p-T153/Y155 antibody showed that phosphorylated TDP-43 was specifically recruited to the nucleoli, suggesting that p-T153/Y155 regulates a previously unappreciated function of TDP-43 in the processing of nucleolar-associated RNA. These findings highlight a new mechanism that regulates TDP-43 function and homeostasis through phosphorylation and, therefore, may contribute to the development of strategies to prevent TDP-43 aggregation and to uncover previously unexplored roles of TDP-43 in cell metabolism.


Subject(s)
DNA-Binding Proteins/metabolism , MAP Kinase Signaling System , HEK293 Cells/chemistry , HeLa Cells , Heat-Shock Response , Humans , MAP Kinase Kinase Kinases/metabolism , Models, Molecular , Phosphorylation , Protein Aggregates , Ribonucleoside Diphosphate Reductase , Tumor Suppressor Proteins/metabolism
4.
Am J Physiol Heart Circ Physiol ; 302(1): H231-43, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22058153

ABSTRACT

The identification of mutations in PTPN11 (encoding the protein tyrosine phosphatase Shp2) in families with congenital heart disease has facilitated mechanistic studies of various cardiovascular defects. However, the roles of normal and mutant Shp2 in the developing heart are still poorly understood. Furthermore, it remains unclear how Shp2 loss-of-function (LOF) mutations cause LEOPARD Syndrome (also termed Noonan Syndrome with multiple lentigines), which is characterized by congenital heart defects such as pulmonary valve stenosis and hypertrophic cardiomyopathy (HCM). In normal hearts, Shp2 controls cardiomyocyte size by regulating signaling through protein kinase B (Akt) and mammalian target of rapamycin (mTOR). We hypothesized that Shp2 LOF mutations dysregulate this pathway, resulting in HCM. For our studies, we chose the Shp2 mutation Q510E, a dominant-negative LOF mutation associated with severe early onset HCM. Newborn mice with cardiomyocyte-specific overexpression of Q510E-Shp2 starting before birth displayed increased cardiomyocyte sizes, heart-to-body weight ratios, interventricular septum thickness, and cardiomyocyte disarray. In 3-mo-old hearts, interstitial fibrosis was detected. Echocardiographically, ventricular walls were thickened and contractile function was depressed. In ventricular tissue samples, signaling through Akt/mTOR was hyperactivated, indicating that the presence of Q510E-Shp2 led to upregulation of this pathway. Importantly, rapamycin treatment started shortly after birth rescued the Q510E-Shp2-induced phenotype in vivo. If rapamycin was started at 6 wk of age, HCM was also ameliorated. We also generated a second mouse model in which cardiomyocyte-specific Q510E-Shp2 overexpression started after birth. In contrast to the first model, these mice did not develop HCM. In summary, our studies establish a role for mTOR signaling in HCM caused by Q510E-Shp2. Q510E-Shp2 overexpression in the cardiomyocyte population alone was sufficient to induce the phenotype. Furthermore, the pathomechanism was triggered pre- but not postnatally. However, postnatal rapamycin treatment could still reverse already established HCM, which may have important therapeutic implications.


Subject(s)
Cardiomyopathy, Hypertrophic/enzymology , Mutation , Myocytes, Cardiac/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Age Factors , Aging/genetics , Aging/metabolism , Animals , Animals, Newborn , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cardiomyopathy, Hypertrophic/prevention & control , Cell Size , Cells, Cultured , Disease Models, Animal , Fibrosis , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transfection , Ventricular Function, Left
5.
Nat Genet ; 54(9): 1390-1405, 2022 09.
Article in English | MEDLINE | ID: mdl-35995947

ABSTRACT

Pancreatic ductal adenocarcinoma is a lethal disease with limited treatment options and poor survival. We studied 83 spatial samples from 31 patients (11 treatment-naïve and 20 treated) using single-cell/nucleus RNA sequencing, bulk-proteogenomics, spatial transcriptomics and cellular imaging. Subpopulations of tumor cells exhibited signatures of proliferation, KRAS signaling, cell stress and epithelial-to-mesenchymal transition. Mapping mutations and copy number events distinguished tumor populations from normal and transitional cells, including acinar-to-ductal metaplasia and pancreatic intraepithelial neoplasia. Pathology-assisted deconvolution of spatial transcriptomic data identified tumor and transitional subpopulations with distinct histological features. We showed coordinated expression of TIGIT in exhausted and regulatory T cells and Nectin in tumor cells. Chemo-resistant samples contain a threefold enrichment of inflammatory cancer-associated fibroblasts that upregulate metallothioneins. Our study reveals a deeper understanding of the intricate substructure of pancreatic ductal adenocarcinoma tumors that could help improve therapy for patients with this disease.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/genetics , Humans , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/genetics , Pancreatic Neoplasms
6.
Endocr Connect ; 5(2): 47-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26830329

ABSTRACT

Follicular thyroid cancer (FTC) is the second most common type of thyroid cancers. In order to develop more effective personalized therapies, it is necessary to thoroughly evaluate patient-derived cell lines in in vivo preclinical models before using them to test new, targeted therapies. This study evaluates the tumorigenic and metastatic potential of a panel of three human FTC cell lines (WRO, FTC-238, and TT1609-CO2) with defined genetic mutations in two in vivo murine models: an orthotopic thyroid cancer model to study tumor progression and a tail vein injection model to study metastasis. All cell lines developed tumors in the orthotopic model, with take rates of 100%. Notably, WRO-derived tumors grew two to four times faster than tumors arising from the FTC-238 and TT2609-CO2 cell lines. These results mirrored those of a tail vein injection model for lung metastasis: one hundred percent of mice injected with WRO cells in the tail vein exhibited aggressive growth of bilateral lung metastases within 35 days. In contrast, tail vein injection of FTC-238 or TT2609-CO2 cells did not result in lung metastasis. Together, our work demonstrates that these human FTC cell lines display highly varied tumorigenic and metastatic potential in vivo with WRO being the most aggressive cell line in both orthotopic and lung metastasis models. This information will be valuable when selecting cell lines for preclinical drug testing.

7.
J Clin Endocrinol Metab ; 100(2): E232-42, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25423568

ABSTRACT

CONTEXT: Anaplastic thyroid carcinoma (ATC) is one of the most deadly human malignancies. It is 99% lethal, and patients have a median survival of only 6 months after diagnosis. Despite these grim statistics, the mechanism underlying the tumorigenic capability of ATC cells is unclear. OBJECTIVE: S100A8 and S100A9 proteins have emerged as critical mediators in cancer. The aim was to investigate the expression and function of S100A8 and S100A9 in ATC and the mechanisms involved. DESIGN: We determined the expression of S100A8 and S100A9 in human ATC by gene array analysis and immunohistochemistry. Using RNAi-mediated stable gene knockdown in human ATC cell lines and bioluminescent imaging of orthotopic and lung metastasis mouse models of human ATC, we investigated the effects of S100A8 and S100A9 on tumorigenesis and metastasis. RESULTS: We demonstrated that S100A8 and S100A9 were overexpressed in ATC but not in other types of thyroid carcinomas. In vivo analysis in mice using ATC cells that had S100A8 knocked down revealed reduced tumor growth and lung metastasis, as well as significantly prolonged animal survival. Mechanistic investigations showed that S100A8 promotes ATC cell proliferation through an interaction with RAGE, which activates the p38, ERK1/2 and JNK signaling pathways in the tumor cells. CONCLUSIONS: These findings establish a novel role for S100A8 in the promoting and enhancing of ATC progression. They further suggest that the inhibition of S100A8 could represent a relevant therapeutic target, with the potential of enabling a more effective treatment path for this deadly disease.


Subject(s)
Calgranulin A/metabolism , Signal Transduction/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Animals , Apoptosis/genetics , Calgranulin A/genetics , Calgranulin B/genetics , Calgranulin B/metabolism , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Humans , Mice , Phosphorylation , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
8.
Thyroid ; 24(7): 1134-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24684373

ABSTRACT

BACKGROUND: We have previously demonstrated that thyrospheres derived from human anaplastic thyroid cancer (ATC) cell lines can reconstitute and sustain tumor growth in vivo. The aim of this study was to use luciferase-expressing thyrospheres to establish a clinically relevant mouse model of ATC that allows noninvasive and sensitive monitoring of tumor progression. METHODS: Two human ATC cell lines stably transfected with a firefly luciferase gene were used to generate thyrospheres under stem cell culture conditions. Cells were orthotopically implanted into the thyroids of immunodeficient NOD/SCIDIl2rg-/- mice to initiate tumors. Tumor progression and metastasis were evaluated by bioluminescent imaging weekly as well as histologic analysis postmortem. RESULTS: We show that only 100 thyrosphere cells are needed for tumor development, and that tumors can be monitored with bioluminescent imaging as early as 7-14 days after implantation. Subsequent histologic evaluation of tissue sections confirmed characteristics of high-grade malignant neoplasms. CONCLUSIONS: This approach offers rapid and highly sensitive noninvasive detection options for the preclinical assessment of novel ATC therapeutics in vivo.


Subject(s)
Carcinogenesis , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Inbred NOD
9.
PLoS One ; 8(5): e65095, 2013.
Article in English | MEDLINE | ID: mdl-23724124

ABSTRACT

Emerging evidence suggests cancer stem cells (CSCs) may initiate new tumors in anaplastic thyroid carcinoma (ATC), one of the most aggressive solid tumors in humans. However, the involvement of CSCs in human tumorigenesis has not been previously studied in authenticated ATC cell lines. Here we demonstrate a functional role of CSCs in four new validated human ATC cell lines (THJ-11T, THJ-16T, THJ-21T and THJ-29T). We identified and enriched CSCs using a spheroid-forming assay. About 3 to 9% of cells from four ATC cell lines formed thyrospheres. The thyrospheres expressed the stem cell markers NANOG and Oct4 and possessed the ability to self-renew. Injection of these thyrospheres into the thyroids of NOD/SCID Il2rg-/- mice resulted in the formation of metastatic tumors that recapitulated the clinical features of human ATC. To our knowledge, this is the first in vivo characterization of thyroid CSCs using validated human ATC cell lines. The availability of disease-specific thyrospheres and our orthotopic tumor models will enable the elucidation of disease mechanisms and the environmental niche of CSCs. They may also be useful for preclinical therapeutic screening and for monitoring the effects of biological therapies on ATC.


Subject(s)
Lung Neoplasms/secondary , Neoplastic Stem Cells/pathology , Thyroid Neoplasms/pathology , Animals , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Clone Cells , Disease Models, Animal , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Phenotype , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/drug therapy , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL