55 results on '"Urrutia RA"'
Search Results
2. Cancer-associated polybromo-1 bromodomain 4 missense variants variably impact bromodomain ligand binding and cell growth suppression.
- Author
-
Bursch KL, Goetz CJ, Jiao G, Nuñez R, Olp MD, Dhiman A, Khurana M, Zimmermann MT, Urrutia RA, Dykhuizen EC, and Smith BC
- Subjects
- Humans, Cell Proliferation, Ligands, Nuclear Proteins genetics, Nuclear Proteins metabolism, Nuclear Proteins chemistry, Protein Binding, Models, Molecular, Protein Structure, Tertiary, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins chemistry, Mutation, Missense, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Protein Domains, Transcription Factors genetics, Transcription Factors metabolism, Transcription Factors chemistry
- Abstract
The polybromo, brahma-related gene 1-associated factors (PBAF) chromatin remodeling complex subunit polybromo-1 (PBRM1) contains six bromodomains that recognize and bind acetylated lysine residues on histone tails and other nuclear proteins. PBRM1 bromodomains thus provide a link between epigenetic posttranslational modifications and PBAF modulation of chromatin accessibility and transcription. As a putative tumor suppressor in several cancers, PBRM1 protein expression is often abrogated by truncations and deletions. However, ∼33% of PBRM1 mutations in cancer are missense and cluster within its bromodomains. Such mutations may generate full-length PBRM1 variant proteins with undetermined structural and functional characteristics. Here, we employed computational, biophysical, and cellular assays to interrogate the effects of PBRM1 bromodomain missense variants on bromodomain stability and function. Since mutations in the fourth bromodomain of PBRM1 (PBRM1-BD4) comprise nearly 20% of all cancer-associated PBRM1 missense mutations, we focused our analysis on PBRM1-BD4 missense protein variants. Selecting 16 potentially deleterious PBRM1-BD4 missense protein variants for further study based on high residue mutational frequency and/or conservation, we show that cancer-associated PBRM1-BD4 missense variants exhibit varied bromodomain stability and ability to bind acetylated histones. Our results demonstrate the effectiveness of identifying the unique impacts of individual PBRM1-BD4 missense variants on protein structure and function, based on affected residue location within the bromodomain. This knowledge provides a foundation for drawing correlations between specific cancer-associated PBRM1 missense variants and distinct alterations in PBRM1 function, informing future cancer personalized medicine approaches., Competing Interests: Conflict of interest The authors declare they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. An EMR-Based Approach to Determine Frequency, Prescribing Pattern, and Characteristics of Patients Receiving Drugs with Pharmacogenomic Guidelines.
- Author
-
MacKinnon GE 3rd, Mills M, Stoddard A, Urrutia RA, and Broeckel U
- Abstract
(1) Background: This retrospective analysis utilizing electronic medical record (EMR) data from a tertiary integrated health system sought to identify patients and prescribers who would benefit from pharmacogenomic (PGx) testing based on Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines. (2) Methods: EMR data from a clinical research data warehouse were analyzed from 845,518 patients that had an encounter between 2015 and 2019 at an academic medical center. Data were collected for 42 commercially available drugs with 52 evidence-based PGx guidelines from CPIC. Provider data were obtained through the EMR linked by specialty via national provider identification (NPI) number. (3) Results: A total of 845,518 patients had an encounter in the extraction period with 590,526 medication orders processed. A total of 335,849 (56.9%) patients had medication orders represented by CPIC drugs prescribed by 2803 providers, representing 239 distinct medications. (4) Conclusions: The results from this study show that over half of patients were prescribed a CPIC actionable medication from a variety of prescriber specialties. Understanding the magnitude of patients that may benefit from PGx testing, will enable the development of preemptive testing processes, physician support strategies, and pharmacist workflows to optimize outcomes should a PGx service be implemented.
- Published
- 2023
- Full Text
- View/download PDF
4. KLF5 and p53 comprise an incoherent feed-forward loop directing cell-fate decisions following stress.
- Author
-
Yang Y, Bhargava D, Chen X, Zhou T, Dursuk G, Jiang W, Wang J, Zong Z, Katz SI, Lomberk GA, Urrutia RA, and Katz JP
- Subjects
- Humans, Apoptosis genetics, Cell Differentiation, Oxidative Stress, Epithelial Cells, Kruppel-Like Transcription Factors genetics, Tumor Suppressor Protein p53 genetics
- Abstract
In response to stress, cells make a critical decision to arrest or undergo apoptosis, mediated in large part by the tumor suppressor p53. Yet the mechanisms of these cell fate decisions remain largely unknown, particularly in normal cells. Here, we define an incoherent feed-forward loop in non-transformed human squamous epithelial cells involving p53 and the zinc-finger transcription factor KLF5 that dictates responses to differing levels of cellular stress from UV irradiation or oxidative stress. In normal unstressed human squamous epithelial cells, KLF5 complexes with SIN3A and HDAC2 repress TP53, allowing cells to proliferate. With moderate stress, this complex is disrupted, and TP53 is induced; KLF5 then acts as a molecular switch for p53 function by transactivating AKT1 and AKT3, which direct cells toward survival. By contrast, severe stress results in KLF5 loss, such that AKT1 and AKT3 are not induced, and cells preferentially undergo apoptosis. Thus, in human squamous epithelial cells, KLF5 gates the response to UV or oxidative stress to determine the p53 output of growth arrest or apoptosis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
5. Impact of integrated translational research on clinical exome sequencing.
- Author
-
Klee EW, Cousin MA, Pinto E Vairo F, Morales-Rosado JA, Macke EL, Jenkinson WG, Ferrer A, Schultz-Rogers LE, Olson RJ, Oliver GR, Sigafoos AN, Schwab TL, Zimmermann MT, Urrutia RA, Kaiwar C, Gupta A, Blackburn PR, Boczek NJ, Prochnow CA, Lowy RJ, Mulvihill LA, McAllister TM, Aoudia SL, Kruisselbrink TM, Gunderson LB, Kemppainen JL, Fisher LJ, Tarnowski JM, Hager MM, Kroc SA, Bertsch NL, Agre KE, Jackson JL, Macklin-Mantia SK, Murphree MI, Rust LM, Summer Bolster JM, Beck SA, Atwal PS, Ellingson MS, Barnett SS, Rasmussen KJ, Lahner CA, Niu Z, Hasadsri L, Ferber MJ, Marcou CA, Clark KJ, Pichurin PN, Deyle DR, Morava-Kozicz E, Gavrilova RH, Dhamija R, Wierenga KJ, Lanpher BC, Babovic-Vuksanovic D, Farrugia G, Schimmenti LA, Stewart AK, and Lazaridis KN
- Published
- 2023
- Full Text
- View/download PDF
6. Effects of Subnormothermic Regulated Hepatic Reperfusion on Mitochondrial and Transcriptomic Profiles in a Porcine Model.
- Author
-
Kim J, Zimmerman MA, Shin WY, Boettcher BT, Lee JS, Park JI, Ali M, Yang M, Mishra J, Hagen CE, McGraw JE, Mathison A, Woehlck HJ, Lomberk G, Camara AKS, Urrutia RA, Stowe DF, and Hong JC
- Subjects
- Swine, Animals, Liver pathology, Reperfusion, Ischemia, Necrosis metabolism, Necrosis pathology, Transcriptome, Organ Preservation methods
- Abstract
Objective: We sought to investigate the biological effects of pre-reperfusion treatments of the liver after warm and cold ischemic injuries in a porcine donation after circulatory death model., Summary of Background Data: Donation after circulatory death represents a severe form of liver ischemia and reperfusion injury that has a profound impact on graft function after liver transplantation., Methods: Twenty donor pig livers underwent 60 minutes of in situ warm ischemia after circulatory arrest and 120 minutes of cold static preservation prior to simulated transplantation using an ex vivo perfusion machine. Four reperfusion treatments were compared: Control-Normothermic (N), Control- Subnormothermic (S), regulated hepatic reperfusion (RHR)-N, and RHR-S (n = 5 each). The biochemical, metabolic, and transcriptomic profiles, as well as mitochondrial function were analyzed., Results: Compared to the other groups, RHR-S treated group showed significantly lower post-reperfusion aspartate aminotransferase levels in the reperfusion effluent and histologic findings of hepatocyte viability and lesser degree of congestion and necrosis. RHR-S resulted in a significantly higher mitochondrial respiratory control index and calcium retention capacity. Transcriptomic profile analysis showed that treatment with RHR-S activated cell survival and viability, cellular homeostasis as well as other biological functions involved in tissue repair such as cytoskeleton or cytoplasm organization, cell migration, transcription, and microtubule dynamics. Furthermore, RHR-S inhibited organismal death, morbidity and mortality, necrosis, and apoptosis., Conclusion: Subnormothermic RHR mitigates IRI and preserves hepatic mitochondrial function after warm and cold hepatic ischemia. This organ resuscitative therapy may also trigger the activation of protective genes against IRI. Sub- normothermic RHR has potential applicability to clinical liver transplantation., Competing Interests: The authors report no conflicts of interest., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. A GATA6-centred gene regulatory network involving HNFs and ΔNp63 controls plasticity and immune escape in pancreatic cancer.
- Author
-
Kloesch B, Ionasz V, Paliwal S, Hruschka N, Martinez de Villarreal J, Öllinger R, Mueller S, Dienes HP, Schindl M, Gruber ES, Stift J, Herndler-Brandstetter D, Lomberk GA, Seidler B, Saur D, Rad R, Urrutia RA, Real FX, and Martinelli P
- Subjects
- Animals, GATA6 Transcription Factor genetics, GATA6 Transcription Factor metabolism, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks, Humans, Mice, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Objective: Molecular taxonomy of tumours is the foundation of personalised medicine and is becoming of paramount importance for therapeutic purposes. Four transcriptomics-based classification systems of pancreatic ductal adenocarcinoma (PDAC) exist, which consistently identified a subtype of highly aggressive PDACs with basal-like features, including ΔNp63 expression and loss of the epithelial master regulator GATA6. We investigated the precise molecular events driving PDAC progression and the emergence of the basal programme., Design: We combined the analysis of patient-derived transcriptomics datasets and tissue samples with mechanistic experiments using a novel dual-recombinase mouse model for Gata6 deletion at late stages of KRas
G12D -driven pancreatic tumorigenesis (Gata6LateKO )., Results: This comprehensive human-to-mouse approach showed that GATA6 loss is necessary, but not sufficient, for the expression of ΔNp63 and the basal programme in patients and in mice. The concomitant loss of HNF1A and HNF4A, likely through epigenetic silencing, is required for the full phenotype switch. Moreover, Gata6 deletion in mice dramatically increased the metastatic rate, with a propensity for lung metastases. Through RNA-Seq analysis of primary cells isolated from mouse tumours, we show that Gata6 inhibits tumour cell plasticity and immune evasion, consistent with patient-derived data, suggesting that GATA6 works as a barrier for acquiring the fully developed basal and metastatic phenotype., Conclusions: Our work provides both a mechanistic molecular link between the basal phenotype and metastasis and a valuable preclinical tool to investigate the most aggressive subtype of PDAC. These data, therefore, are important for understanding the pathobiological features underlying the heterogeneity of pancreatic cancer in both mice and human., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2022
- Full Text
- View/download PDF
8. Microglia Influence Neurofilament Deposition in ALS iPSC-Derived Motor Neurons.
- Author
-
Allison RL, Adelman JW, Abrudan J, Urrutia RA, Zimmermann MT, Mathison AJ, and Ebert AD
- Subjects
- Culture Media, Conditioned, Humans, Intermediate Filaments metabolism, Male, Microglia metabolism, Motor Neurons metabolism, Amyotrophic Lateral Sclerosis metabolism, Induced Pluripotent Stem Cells metabolism, Neurodegenerative Diseases metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which upper and lower motor neuron loss is the primary phenotype, leading to muscle weakness and wasting, respiratory failure, and death. Although a portion of ALS cases are linked to one of over 50 unique genes, the vast majority of cases are sporadic in nature. However, the mechanisms underlying the motor neuron loss in either familial or sporadic ALS are not entirely clear. Here, we used induced pluripotent stem cells derived from a set of identical twin brothers discordant for ALS to assess the role of astrocytes and microglia on the expression and accumulation of neurofilament proteins in motor neurons. We found that motor neurons derived from the affected twin which exhibited increased transcript levels of all three neurofilament isoforms and increased expression of phosphorylated neurofilament puncta. We further found that treatment of the motor neurons with astrocyte-conditioned medium and microglial-conditioned medium significantly impacted neurofilament deposition. Together, these data suggest that glial-secreted factors can alter neurofilament pathology in ALS iPSC-derived motor neurons.
- Published
- 2022
- Full Text
- View/download PDF
9. Impact of integrated translational research on clinical exome sequencing.
- Author
-
Klee EW, Cousin MA, Pinto E Vairo F, Morales-Rosado JA, Macke EL, Jenkinson WG, Ferrer A, Schultz-Rogers LE, Olson RJ, Oliver GR, Sigafoos AN, Schwab TL, Zimmermann MT, Urrutia RA, Kaiwar C, Gupta A, Blackburn PR, Boczek NJ, Prochnow CA, Lowy RJ, Mulvihill LA, McAllister TM, Aoudia SL, Kruisselbrink TM, Gunderson LB, Kemppainen JL, Fisher LJ, Tarnowski JM, Hager MM, Kroc SA, Bertsch NL, Agre KE, Jackson JL, Macklin-Mantia SK, Murphree MI, Rust LM, Summer Bolster JM, Beck SA, Atwal PS, Ellingson MS, Barnett SS, Rasmussen KJ, Lahner CA, Niu Z, Hasadsri L, Ferber MJ, Marcou CA, Clark KJ, Pichurin PN, Deyle DR, Morava-Kozicz E, Gavrilova RH, Dhamija R, Wierenga KJ, Lanpher BC, Babovic-Vuksanovic D, Farrugia G, Schimmenti LA, Stewart AK, and Lazaridis KN
- Subjects
- Genetic Testing, Humans, Phenotype, Translational Research, Biomedical, Exome Sequencing, Exome genetics, Undiagnosed Diseases
- Abstract
Purpose: Exome sequencing often identifies pathogenic genetic variants in patients with undiagnosed diseases. Nevertheless, frequent findings of variants of uncertain significance necessitate additional efforts to establish causality before reaching a conclusive diagnosis. To provide comprehensive genomic testing to patients with undiagnosed disease, we established an Individualized Medicine Clinic, which offered clinical exome testing and included a Translational Omics Program (TOP) that provided variant curation, research activities, or research exome sequencing., Methods: From 2012 to 2018, 1101 unselected patients with undiagnosed diseases received exome testing. Outcomes were reviewed to assess impact of the TOP and patient characteristics on diagnostic rates through descriptive and multivariate analyses., Results: The overall diagnostic yield was 24.9% (274 of 1101 patients), with 174 (15.8% of 1101) diagnosed on the basis of clinical exome sequencing alone. Four hundred twenty-three patients with nondiagnostic or without access to clinical exome sequencing were evaluated by the TOP, with 100 (9% of 1101) patients receiving a diagnosis, accounting for 36.5% of the diagnostic yield. The identification of a genetic diagnosis was influenced by the age at time of testing and the disease phenotype of the patient., Conclusion: Integration of translational research activities into clinical practice of a tertiary medical center can significantly increase the diagnostic yield of patients with undiagnosed disease.
- Published
- 2021
- Full Text
- View/download PDF
10. Inorganic arsenic promotes luminal to basal transition and metastasis of breast cancer.
- Author
-
Danes JM, de Abreu ALP, Kerketta R, Huang Y, Palma FR, Gantner BN, Mathison AJ, Urrutia RA, and Bonini MG
- Subjects
- Animals, Breast metabolism, Breast Neoplasms metabolism, Cell Line, Tumor, Estrogen Receptor alpha metabolism, Estrogens metabolism, Female, Humans, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Signal Transduction drug effects, Arsenic physiology, Breast drug effects, Breast pathology, Breast Neoplasms chemically induced, Breast Neoplasms pathology
- Abstract
Inorganic arsenic (iAs/As
2 O3 2- ) is an environmental toxicant found in watersheds around the world including in densely populated areas. iAs is a class I carcinogen known to target the skin, lungs, bladder, and digestive organs, but its role as a primary breast carcinogen remains controversial. Here, we examined a different possibility: that exposure to iAs promotes the transition of well-differentiated epithelial breast cancer cells characterized by estrogen and progesterone receptor expression (ER+/PR+), to more basal phenotypes characterized by active proliferation, and propensity to metastasis in vivo. Our results indicate two clear phenotypic responses to low-level iAs that depend on the duration of the exposure. Short-term pulses of iAs activate ER signaling, consistent with its reported pseudo-estrogen activity, but longer-term, chronic treatments for over 6 months suppresses both ER and PR expression and signaling. In fact, washout of these chronically exposed cells for up to 1 month failed to fully reverse the transcriptional and phenotypic effects of prolonged treatments, indicating durable changes in cellular physiologic identity. RNA-seq studies found that chronic iAs drives the transition toward more basal phenotypes characterized by impaired hormone receptor signaling despite the conservation of estrogen receptor expression. Because treatments for breast cancer patients are largely designed based on the detection of hormone receptor expression, our results suggest greater scrutiny of ER+ cancers in patients exposed to iAs, because these tumors may spawn more aggressive phenotypes than unexposed ER+ tumors, in particular, basal subtypes that tend to develop therapy resistance and metastasis., (© 2020 Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
11. Variant anatomy of the biliary system as a cause of pancreatic and peri-ampullary cancers.
- Author
-
Muraki T, Reid MD, Pehlivanoglu B, Gonzalez RS, Sekhar A, Memis B, Xue Y, Cheng J, Jang KT, Mittal P, Cardona K, Kooby DA, Maithel S, Sarmiento JM, El-Rayes B, Lomberk G, Urrutia RA, Christians K, Tsai S, Evans DB, and Adsay V
- Subjects
- Bile Ducts, Intrahepatic, Humans, Ampulla of Vater, Bile Duct Neoplasms diagnostic imaging, Bile Duct Neoplasms surgery, Common Bile Duct Neoplasms, Duodenal Neoplasms, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms surgery
- Abstract
Background: The cause of most pancreatic and periampullary cancers (PAC) is unknown. Recently, anatomic variations such as pancreatobiliary maljunction have been recognized as risk factors, similar to Barrett-related gastro-esophageal cancers., Methods: Pre-operative MRI from 860 pancreatic/biliary resections, including 322 PACs, were evaluated for low-union (cystic duct joining the common hepatic duct inside of the pancreas or within 5 mm of the pancreatic border) RESULTS: Low-union, seen <10% of the population, was present in 44% of PACs (73% distal bile duct/cholangiocarcinoma, 42% pancreatic head, and 34% ampullary). It was significantly lower(11%) in conditions without connection to the ductal system (thus not exposed to the ductal/biliary tract contents), namely mucinous cystic neoplasms and intrahepatic cholangiocarcinomas(p < 0.0001). Intra-pancreatic type low-union was seen in 16% of PACs versus 2% of controls(p < 0.0001)., Discussion: This study establishes an association between low-union and PACs, and points to an anatomy-induced chemical/bilious carcinogenesis. This may explain why most pancreas cancers are in the head. It is possible that the same chemical milieu, caused by conditions other than low-union/insertion, may also play a role in the remaining half of PACs. This opens various treatment opportunities including milieu modifications (chemoprevention), focused screening of at-risk patients, and early detection with possible corrective actions., (Copyright © 2020 International Hepato-Pancreato-Biliary Association Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. Targeting the CBP/β-Catenin Interaction to Suppress Activation of Cancer-Promoting Pancreatic Stellate Cells.
- Author
-
Che M, Kweon SM, Teo JL, Yuan YC, Melstrom LG, Waldron RT, Lugea A, Urrutia RA, Pandol SJ, and Lai KKY
- Abstract
Background: Although cyclic AMP-response element binding protein-binding protein (CBP)/β-catenin signaling is known to promote proliferation and fibrosis in various organ systems, its role in the activation of pancreatic stellate cells (PSCs), the key effector cells of desmoplasia in pancreatic cancer and fibrosis in chronic pancreatitis, is largely unknown., Methods: To investigate the role of the CBP/β-catenin signaling pathway in the activation of PSCs, we have treated mouse and human PSCs with the small molecule specific CBP/β-catenin antagonist ICG-001 and examined the effects of treatment on parameters of activation., Results: We report for the first time that CBP/β-catenin antagonism suppresses activation of PSCs as evidenced by their decreased proliferation, down-regulation of "activation" markers, e.g., α-smooth muscle actin (α-SMA/Acta2), collagen type I alpha 1 (Col1a1), Prolyl 4-hydroxylase, and Survivin, up-regulation of peroxisome proliferator activated receptor gamma (Ppar-γ) which is associated with quiescence, and reduced migration; additionally, CBP/β-catenin antagonism also suppresses PSC-induced migration of cancer cells., Conclusion: CBP/β-catenin antagonism represents a novel therapeutic strategy for suppressing PSC activation and may be effective at countering PSC promotion of pancreatic cancer.
- Published
- 2020
- Full Text
- View/download PDF
13. Single-cell transcriptome reveals the novel role of T-bet in suppressing the immature NK gene signature.
- Author
-
Yang C, Siebert JR, Burns R, Zheng Y, Mei A, Bonacci B, Wang D, Urrutia RA, Riese MJ, Rao S, Carlson KS, Thakar MS, and Malarkannan S
- Subjects
- Animals, Bone Marrow Cells immunology, Cluster Analysis, Forkhead Box Protein O1 genetics, Forkhead Box Protein O1 metabolism, Gene Expression Regulation, Genotype, Killer Cells, Natural immunology, Mechanistic Target of Rapamycin Complex 2 deficiency, Mechanistic Target of Rapamycin Complex 2 genetics, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Rapamycin-Insensitive Companion of mTOR Protein deficiency, Rapamycin-Insensitive Companion of mTOR Protein genetics, Regulatory-Associated Protein of mTOR deficiency, Regulatory-Associated Protein of mTOR genetics, T-Box Domain Proteins metabolism, Bone Marrow Cells metabolism, Gene Expression Profiling, Killer Cells, Natural metabolism, Machine Learning, RNA-Seq, Single-Cell Analysis, T-Box Domain Proteins genetics, Transcriptome
- Abstract
The transcriptional activation and repression during NK cell ontology are poorly understood. Here, using single-cell RNA-sequencing, we reveal a novel role for T-bet in suppressing the immature gene signature during murine NK cell development. Based on transcriptome, we identified five distinct NK cell clusters and define their relative developmental maturity in the bone marrow. Transcriptome-based machine-learning classifiers revealed that half of the mTORC2-deficient NK cells belongs to the least mature NK cluster. Mechanistically, loss of mTORC2 results in an increased expression of signature genes representing immature NK cells. Since mTORC2 regulates the expression of T-bet through Akt
S473 -FoxO1 axis, we further characterized the T-bet-deficient NK cells and found an augmented immature transcriptomic signature. Moreover, deletion of Foxo1 restores the expression of T-bet and corrects the abnormal expression of immature NK genes. Collectively, our study reveals a novel role for mTORC2-AktS473 -FoxO1-T-bet axis in suppressing the transcriptional signature of immature NK cells., Competing Interests: CY, JS, RB, YZ, AM, BB, DW, RU, MR, SR, KC, MT, SM No competing interests declared, (© 2020, Yang et al.)- Published
- 2020
- Full Text
- View/download PDF
14. Genetic variants in DGAT1 cause diverse clinical presentations of malnutrition through a specific molecular mechanism.
- Author
-
Gupta A, Dsouza NR, Zarate YA, Lombardo R, Hopkin R, Linehan AR, Simpson J, McCarrier J, Agre KE, Gavrilova RH, Stephens MC, Grothe RM, Monaghan KG, Xie Y, Basel D, Urrutia RA, Cole CR, Klee EW, and Zimmermann MT
- Subjects
- Diarrhea diet therapy, Female, Humans, Infant, Infant, Newborn, Male, Malnutrition diet therapy, Mutation, Exome Sequencing, Diacylglycerol O-Acyltransferase genetics, Diarrhea genetics, Malnutrition genetics
- Abstract
Background: DGAT1, a gene encoding a protein involved in lipid metabolism, has been recently implicated in causing a rare nutritional and digestive disease presenting as Congenital Diarrheal Disorder (CDD). Genetic causes of malnutrition can be classified as metabolic disorders, caused by loss of a specific enzyme's function. However, disease driven by genetic variants in lipid metabolism genes is not well understood, and additional information is needed to better understand these effects., Methods: We gathered a multi-institutional cohort of undiagnosed patients with a constellation of phenotypes presenting as malnutrition and metal ion dysregulation. Clinical Whole Exome Sequencing (WES) was performed on four patients and their unaffected parents. We prioritized genetic variants based on multiple criteria including population allele frequency and presumed inheritance pattern, and identified a candidate gene. Computational modeling was used to investigate if the altered amino acids are likely to result in a dysfunctional enzyme., Results: We identified a multi-institutional cohort of patients presenting with malnutrition-like symptoms and likely pathogenic genomic variants within DGAT1. Multiple approaches were used to profile the effect these variants have on protein structure and function. Laboratory and nutritional intervention studies showed rapid and robust patient responses., Conclusions: This report adds on to the database for existing mutations known within DGAT1, a gene recently implicated with CDD, and also expands its clinical spectrum. Identification of these DGAT1 mutations by WES has allowed for changes in the patients' nutritional rehabilitation, reversed growth failure and enabled them to be weaned off of total parenteral nutrition (TPN)., Competing Interests: Declaration of competing interest Kristin G. Monaghan and Yili Xie are employees of GeneDx, Inc., a wholly owned subsidiary of OPKO Health, Inc. All other authors declare no conflicts of interest., (Copyright © 2019 Elsevier Masson SAS. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
15. Motor Neuron Generation from iPSCs from Identical Twins Discordant for Amyotrophic Lateral Sclerosis.
- Author
-
Seminary ER, Santarriaga S, Wheeler L, Mejaki M, Abrudan J, Demos W, Zimmermann MT, Urrutia RA, Fee D, Barkhaus PE, and Ebert AD
- Subjects
- Amyotrophic Lateral Sclerosis genetics, Autophagy, Cell Survival, Glutamic Acid metabolism, Humans, Male, Middle Aged, Motor Neurons metabolism, Protein Aggregates, Solubility, Whole Genome Sequencing, Amyotrophic Lateral Sclerosis pathology, Induced Pluripotent Stem Cells pathology, Motor Neurons pathology, Twins, Monozygotic
- Abstract
Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disorder characterized by the loss of the upper and lower motor neurons. Approximately 10% of cases are caused by specific mutations in known genes, with the remaining cases having no known genetic link. As such, sporadic cases have been more difficult to model experimentally. Here, we describe the generation and differentiation of ALS induced pluripotent stem cells reprogrammed from discordant identical twins. Whole genome sequencing revealed no relevant mutations in known ALS-causing genes that differ between the twins. As protein aggregation is found in all ALS patients and is thought to contribute to motor neuron death, we sought to characterize the aggregation phenotype of the sporadic ALS induced pluripotent stem cells (iPSCs). Motor neurons from both twins had high levels of insoluble proteins that commonly aggregate in ALS that did not robustly change in response to exogenous glutamate. In contrast, established genetic ALS iPSC lines demonstrated insolubility in a protein- and genotype-dependent manner. Moreover, whereas the genetic ALS lines failed to induce autophagy after glutamate stress, motor neurons from both twins and independent controls did activate this protective pathway. Together, these data indicate that our unique model of sporadic ALS may provide key insights into disease pathology and highlight potential differences between sporadic and familial ALS., Competing Interests: DF has received personal fees from serving on the Biogen ALS Advisory Board. The authors declare no other conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.
- Published
- 2020
- Full Text
- View/download PDF
16. SOD2 acetylation on lysine 68 promotes stem cell reprogramming in breast cancer.
- Author
-
He C, Danes JM, Hart PC, Zhu Y, Huang Y, de Abreu AL, O'Brien J, Mathison AJ, Tang B, Frasor JM, Wakefield LM, Ganini D, Stauder E, Zielonka J, Gantner BN, Urrutia RA, Gius D, and Bonini MG
- Subjects
- Acetylation, Animals, Basic Helix-Loop-Helix Transcription Factors physiology, Breast Neoplasms metabolism, Cellular Reprogramming, Disease Progression, Female, Heterografts, Humans, Hydrogen Peroxide metabolism, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Mitochondria enzymology, Neoplasm Invasiveness, Neoplasm Proteins chemistry, Protein Processing, Post-Translational, Recombinant Proteins metabolism, Superoxide Dismutase chemistry, Breast Neoplasms pathology, Cell Self Renewal physiology, Neoplasm Proteins physiology, Neoplastic Stem Cells physiology, Superoxide Dismutase physiology
- Abstract
Mitochondrial superoxide dismutase (SOD2) suppresses tumor initiation but promotes invasion and dissemination of tumor cells at later stages of the disease. The mechanism of this functional switch remains poorly defined. Our results indicate that as SOD2 expression increases acetylation of lysine 68 ensues. Acetylated SOD2 promotes hypoxic signaling via increased mitochondrial reactive oxygen species (mtROS). mtROS, in turn, stabilize hypoxia-induced factor 2α (HIF2α), a transcription factor upstream of "stemness" genes such as Oct4, Sox2, and Nanog. In this sense, our findings indicate that SOD2
K68Ac and mtROS are linked to stemness reprogramming in breast cancer cells via HIF2α signaling. Based on these findings we propose that, as tumors evolve, the accumulation of SOD2K68Ac turns on a mitochondrial pathway to stemness that depends on HIF2α and may be relevant for the progression of breast cancer toward poor outcomes., Competing Interests: The authors declare no competing interest.- Published
- 2019
- Full Text
- View/download PDF
17. Molecular characterization of known and novel ACVR1 variants in phenotypes of aberrant ossification.
- Author
-
Gupta A, Zimmermann MT, Wang H, Broski SM, Sigafoos AN, Macklin SK, Urrutia RA, Clark KJ, Atwal PS, Pignolo RJ, and Klee EW
- Subjects
- Adolescent, Adult, Algorithms, Computer Simulation, Female, Humans, Longitudinal Ligaments physiopathology, Male, Molecular Dynamics Simulation, Muscle, Skeletal physiopathology, Mutation genetics, Myositis Ossificans blood, Myositis Ossificans diagnostic imaging, Myositis Ossificans physiopathology, Ossification of Posterior Longitudinal Ligament physiopathology, Ossification, Heterotopic diagnostic imaging, Ossification, Heterotopic physiopathology, Phenotype, Signal Transduction genetics, Smad Proteins genetics, Activin Receptors, Type I genetics, Muscle, Skeletal metabolism, Myositis Ossificans genetics, Ossification of Posterior Longitudinal Ligament genetics, Ossification, Heterotopic genetics
- Abstract
Diffuse idiopathic skeletal hyperostosis (DISH) is a disorder principally characterized by calcification and ossification of spinal ligaments and entheses. Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disabling disorder characterized by progressive ossification of skeletal muscle, fascia, tendons, and ligaments. These conditions manifest phenotypic overlap in the ossification of tendons and ligaments. We describe herein a patient with DISH, exhibiting heterotopic ossification of the posterior longitudinal ligament where clinical whole exome sequencing identified a variant within ACVR1, a gene implicated in FOP. This variant, p.K400E, is a novel variant, not identified previously, and occurs in a highly conserved region across orthologs. We used sequence-based predicative algorithms, molecular modeling, and molecular dynamics simulations, to test the potential for p.K400E to alter the structure and dynamics of ACVR1. We applied the same modeling and simulation methods to established FOP variants, to identify the detailed effects that they have on the ACVR1 protein, as well as to act as positive controls against which the effects of p.K400E could be evaluated. Our in silico molecular analyses support p.K400E as altering the behavior of ACVR1. In addition, functional testing to measure the effect of this variant on BMP-pSMAD 1/5/8 target genes was carried out which revealed this variant to cause increased ID1 and Msx2 expression compared with the wild-type receptor. This analysis supports the potential for the variant of uncertain significance to contribute to the patient's phenotype., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
18. Precision Medicine and Precision Public Health: Academic Education and Community Engagement.
- Author
-
Meurer JR, Whittle JC, Lamb KM, Kosasih MA, Dwinell MR, and Urrutia RA
- Subjects
- Data Collection, Health Education, Humans, Cooperative Behavior, Precision Medicine, Public Health
- Published
- 2019
- Full Text
- View/download PDF
19. Aurora kinase B-phosphorylated HP1α functions in chromosomal instability.
- Author
-
Williams MM, Mathison AJ, Christensen T, Greipp PT, Knutson DL, Klee EW, Zimmermann MT, Iovanna J, Lomberk GA, and Urrutia RA
- Subjects
- Animals, Aurora Kinase B antagonists & inhibitors, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone genetics, Chromosome Aberrations, HeLa Cells, Heterochromatin metabolism, Humans, Kinetochores metabolism, Mice, Mice, Inbred C57BL, Mitosis, Phosphorylation, Phosphoserine metabolism, Protein Binding, Protein Kinase Inhibitors pharmacology, Aurora Kinase B metabolism, Chromosomal Instability, Chromosomal Proteins, Non-Histone metabolism
- Abstract
Heterochromatin Protein 1 α (HP1α) associates with members of the chromosome passenger complex (CPC) during mitosis, at centromeres where it is required for full Aurora Kinase B (AURKB) activity. Conversely, recent reports have identified AURKB as the major kinase responsible for phosphorylation of HP1α at Serine 92 (S92) during mitosis. Thus, the current study was designed to better understand the functional role of this posttranslationally modified form of HP1α. We find that S92-phosphorylated HP1α is generated in cells at early prophase, localizes to centromeres, and associates with regulators of chromosome stability, such as Inner Centromere Protein, INCENP. In mouse embryonic fibroblasts, HP1α knockout alone or reconstituted with a non-phosphorylatable (S92A) HP1α mutant results in mitotic chromosomal instability characterized by the formation of anaphase/telophase chromatin bridges and micronuclei. These effects are rescued by exogenous expression of wild type HP1α or a phosphomimetic (S92D) variant. Thus, the results from the current study extend our knowledge of the role of HP1α in chromosomal stability during mitosis.
- Published
- 2019
- Full Text
- View/download PDF
20. CBX5/G9a/H3K9me-mediated gene repression is essential to fibroblast activation during lung fibrosis.
- Author
-
Ligresti G, Caporarello N, Meridew JA, Jones DL, Tan Q, Choi KM, Haak AJ, Aravamudhan A, Roden AC, Prakash YS, Lomberk G, Urrutia RA, and Tschumperlin DJ
- Subjects
- Animals, Antibiotics, Antineoplastic toxicity, Bleomycin toxicity, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone metabolism, Collagen metabolism, Disease Models, Animal, Epigenesis, Genetic, Fibroblasts pathology, Gene Silencing, Histocompatibility Antigens metabolism, Histone Code genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, Idiopathic Pulmonary Fibrosis metabolism, Idiopathic Pulmonary Fibrosis pathology, Lung cytology, Lung pathology, Mice, Mice, Transgenic, Transforming Growth Factor beta metabolism, Chromosomal Proteins, Non-Histone genetics, Fibroblasts metabolism, Histocompatibility Antigens genetics, Histone-Lysine N-Methyltransferase genetics, Idiopathic Pulmonary Fibrosis genetics, Lung metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics
- Abstract
Pulmonary fibrosis is a devastating disease characterized by accumulation of activated fibroblasts and scarring in the lung. While fibroblast activation in physiological wound repair reverses spontaneously, fibroblast activation in fibrosis is aberrantly sustained. Here we identified histone 3 lysine 9 methylation (H3K9me) as a critical epigenetic modification that sustains fibroblast activation by repressing the transcription of genes essential to returning lung fibroblasts to an inactive state. We show that the histone methyltransferase G9a (EHMT2) and chromobox homolog 5 (CBX5, also known as HP1α), which deposit H3K9me marks and assemble an associated repressor complex respectively, are essential to initiation and maintenance of fibroblast activation specifically through epigenetic repression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha gene (PPARGC1A, encoding PGC1α). Both TGFβ and increased matrix stiffness potently inhibit PGC1α expression in lung fibroblasts through engagement of the CBX5/G9a pathway. Inhibition of CBX5/G9a pathway in fibroblasts elevates PGC1α, attenuates TGFβ- and matrix stiffness-promoted H3K9 methylation, and reduces collagen accumulation in the lungs following bleomycin injury. Our results demonstrate that epigenetic silencing mediated by H3K9 methylation is essential for both biochemical and biomechanical fibroblast activation, and that targeting this epigenetic pathway may provide therapeutic benefit by returning lung fibroblasts to quiescence.
- Published
- 2019
- Full Text
- View/download PDF
21. Disruption of FOXP3-EZH2 Interaction Represents a Pathobiological Mechanism in Intestinal Inflammation.
- Author
-
Bamidele AO, Svingen PA, Sagstetter MR, Sarmento OF, Gonzalez M, Braga Neto MB, Kugathasan S, Lomberk G, Urrutia RA, and Faubion WA Jr
- Subjects
- Adult, Animals, Cell Nucleus metabolism, Cell Separation, Co-Repressor Proteins metabolism, Female, Humans, Inflammation immunology, Inflammatory Bowel Diseases immunology, Inflammatory Bowel Diseases pathology, Interleukin-6 metabolism, Janus Kinases metabolism, Jurkat Cells, Male, Mice, Inbred C57BL, Middle Aged, Mutation genetics, Phosphorylation, Phosphotyrosine metabolism, Polycomb Repressive Complex 2 metabolism, Protein Binding, STAT3 Transcription Factor metabolism, Signal Transduction, T-Lymphocytes, Regulatory metabolism, Enhancer of Zeste Homolog 2 Protein metabolism, Forkhead Transcription Factors metabolism, Inflammation metabolism, Inflammation pathology, Intestines pathology
- Abstract
Background & Aims: Forkhead box protein 3 (FOXP3)
+ regulatory T cell (Treg) dysfunction is associated with autoimmune diseases; however, the mechanisms responsible for inflammatory bowel disease pathophysiology are poorly understood. Here, we tested the hypothesis that a physical interaction between transcription factor FOXP3 and the epigenetic enzyme enhancer of zeste homolog 2 (EZH2) is essential for gene co-repressive function., Methods: Human FOXP3 mutations clinically relevant to intestinal inflammation were generated by site-directed mutagenesis. T lymphocytes were isolated from mice, human blood, and lamina propria of Crohn's disease (CD) patients and non-CD controls. We performed proximity ligation or a co-immunoprecipitation assay in FOXP3-mutant+ , interleukin 6 (IL6)-treated or CD-CD4+ T cells to assess FOXP3-EZH2 protein interaction. We studied IL2 promoter activity and chromatin state of the interferon γ locus via luciferase reporter and chromatin-immunoprecipitation assays, respectively, in cells expressing FOXP3 mutants., Results: EZH2 binding was abrogated by inflammatory bowel disease-associated FOXP3 cysteine 232 (C232) mutation. The C232 mutant showed impaired repression of IL2 and diminished EZH2-mediated trimethylation of histone 3 at lysine 27 on interferon γ, indicative of compromised Treg physiologic function. Generalizing this mechanism, IL6 impaired FOXP3-EZH2 interaction. IL6-induced effects were reversed by Janus kinase 1/2 inhibition. In lamina propria-derived CD4+ T cells from CD patients, we observed decreased FOXP3-EZH2 interaction., Conclusions: FOXP3-C232 mutation disrupts EZH2 recruitment and gene co-repressive function. The proinflammatory cytokine IL6 abrogates FOXP3-EZH2 interaction. Studies in lesion-derived CD4+ T cells have shown that reduced FOXP3-EZH2 interaction is a molecular feature of CD patients. Destabilized FOXP3-EZH2 protein interaction via diverse mechanisms and consequent Treg abnormality may drive gastrointestinal inflammation.- Published
- 2018
- Full Text
- View/download PDF
22. Co-occurrence of a maternally inherited DNMT3A duplication and a paternally inherited pathogenic variant in EZH2 in a child with growth retardation and severe short stature: atypical Weaver syndrome or evidence of a DNMT3A dosage effect?
- Author
-
Polonis K, Blackburn PR, Urrutia RA, Lomberk GA, Kruisselbrink T, Cousin MA, Boczek NJ, Hoppman NL, Babovic-Vuksanovic D, Klee EW, and Pichurin PN
- Subjects
- Abnormalities, Multiple pathology, Adult, Child, Congenital Hypothyroidism pathology, Craniofacial Abnormalities pathology, DNA Methyltransferase 3A, Female, Gene Dosage, Hand Deformities, Congenital pathology, Humans, Male, Pedigree, Phenotype, Abnormalities, Multiple genetics, Congenital Hypothyroidism genetics, Craniofacial Abnormalities genetics, DNA (Cytosine-5-)-Methyltransferases genetics, Enhancer of Zeste Homolog 2 Protein genetics, Gene Duplication, Hand Deformities, Congenital genetics
- Abstract
Overgrowth syndromes are a clinically heterogeneous group of disorders characterized by localized or generalized tissue overgrowth and varying degrees of developmental and intellectual disability. An expanding list of genes associated with overgrowth syndromes include the histone methyltransferase genes EZH2 and NSD1 , which cause Weaver and Sotos syndrome, respectively, and the DNA methyltransferase ( DNMT3A ) gene that results in Tatton-Brown-Rahman syndrome (TBRS). Here, we describe a 5-year-old female with a paternally inherited pathogenic mutation in EZH2 (c.2050C>T, p.Arg684Cys) and a maternally inherited 505-kb duplication of uncertain significance at 2p23.3 (encompassing five genes, including DNMT3A ) who presented with intrauterine growth restriction, slow postnatal growth, short stature, hypotonia, developmental delay, and neuroblastoma diagnosed at the age of 8 mo. Her father had tall stature, dysmorphic facial features, and intellectual disability consistent with Weaver syndrome, whereas her mother had short stature, cognitive delays, and chronic nonprogressive leukocytosis. It has been previously shown that EZH2 directly controls DNA methylation through physical association with DNMTs, including DNMT3A, with concomitant H3K27 methylation and CpG promoter methylation leading to repression of EZH2 target genes. Interestingly, NSD1 is involved in H3K36 methylation, a mark associated with transcriptional activation, and exhibits exquisite dosage sensitivity leading to overgrowth when deleted and severe undergrowth when duplicated in vivo. Although there is currently no evidence of dosage effects for DNMT3A , the co-occurrence of a duplication involving this gene and a pathogenic alteration in EZH2 in a patient with severe undergrowth is suggestive of a similar paradigm and further study is warranted., (© 2018 Polonis et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
23. Novel NR2F1 variants likely disrupt DNA binding: molecular modeling in two cases, review of published cases, genotype-phenotype correlation, and phenotypic expansion of the Bosch-Boonstra-Schaaf optic atrophy syndrome.
- Author
-
Kaiwar C, Zimmermann MT, Ferber MJ, Niu Z, Urrutia RA, Klee EW, and Babovic-Vuksanovic D
- Subjects
- Adolescent, Autism Spectrum Disorder genetics, Child, Child, Preschool, DNA, DNA-Binding Proteins genetics, Developmental Disabilities genetics, Exome genetics, Female, Genetic Association Studies, Humans, Intellectual Disability genetics, Language Development Disorders genetics, Male, Models, Molecular, Muscle Hypotonia genetics, Mutation genetics, Optic Atrophies, Hereditary metabolism, Optic Atrophy genetics, Seizures genetics, Exome Sequencing, COUP Transcription Factor I genetics, COUP Transcription Factor I metabolism, Optic Atrophies, Hereditary genetics
- Abstract
Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is a recently described autosomal dominant disorder caused by mutations in the NR2F1 gene. There are presently 28 cases of BBSOAS described in the literature. Its common features include developmental delay, intellectual disability, hypotonia, optic nerve atrophy, attention deficit disorder, autism spectrum disorder, seizures, hearing defects, spasticity, and thinning of the corpus callosum. Here we report two unrelated probands with novel, de novo, missense variants in NR2F1 The first is a 14-yr-old male patient with hypotonia, intellectual disability, optic nerve hypoplasia, delayed bone age, short stature, and altered neurotransmitter levels on cerebrospinal fluid testing. The second is a 5-yr-old female with severe developmental delay, motor and speech delay, and repetitive motion behavior. Whole-exome sequencing identified a novel missense NR2F1 variant in each case, Cys86Phe in the DNA-binding domain in Case 1, and a Leu372Pro in the ligand-binding domain in Case 2. The presence of clinical findings compatible with BBSOAS along with structural analysis at atomic resolution using homology-based molecular modeling and molecular dynamic simulations, support the pathogenicity of these variants for BBSOAS. Short stature, abnormal CNS neurotransmitters, and macrocephaly have not been previously reported for this syndrome and may represent a phenotypic expansion of BBSOAS. A review of published cases along with new evidence from this report support genotype-phenotype correlations for this disorder., (© 2017 Kaiwar et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2017
- Full Text
- View/download PDF
24. Resolution of Cochlear Inflammation: Novel Target for Preventing or Ameliorating Drug-, Noise- and Age-related Hearing Loss.
- Author
-
Kalinec GM, Lomberk G, Urrutia RA, and Kalinec F
- Abstract
A significant number of studies support the idea that inflammatory responses are intimately associated with drug-, noise- and age-related hearing loss (DRHL, NRHL and ARHL). Consequently, several clinical strategies aimed at reducing auditory dysfunction by preventing inflammation are currently under intense scrutiny. Inflammation, however, is a normal adaptive response aimed at restoring tissue functionality and homeostasis after infection, tissue injury and even stress under sterile conditions, and suppressing it could have unintended negative consequences. Therefore, an appropriate approach to prevent or ameliorate DRHL, NRHL and ARHL should involve improving the resolution of the inflammatory process in the cochlea rather than inhibiting this phenomenon. The resolution of inflammation is not a passive response but rather an active, highly controlled and coordinated process. Inflammation by itself produces specialized pro-resolving mediators with critical functions, including essential fatty acid derivatives (lipoxins, resolvins, protectins and maresins), proteins and peptides such as annexin A1 and galectins, purines (adenosine), gaseous mediators (NO, H
2 S and CO), as well as neuromodulators like acetylcholine and netrin-1. In this review article, we describe recent advances in the understanding of the resolution phase of inflammation and highlight therapeutic strategies that might be useful in preventing inflammation-induced cochlear damage. In particular, we emphasize beneficial approaches that have been tested in pre-clinical models of inflammatory responses induced by recognized ototoxic drugs such as cisplatin and aminoglycoside antibiotics. Since these studies suggest that improving the resolution process could be useful for the prevention of inflammation-associated diseases in humans, we discuss the potential application of similar strategies to prevent or mitigate DRHL, NRHL and ARHL.- Published
- 2017
- Full Text
- View/download PDF
25. Functional validation reveals the novel missense V419L variant in TGFBR2 associated with Loeys-Dietz syndrome (LDS) impairs canonical TGF-β signaling.
- Author
-
Cousin MA, Zimmermann MT, Mathison AJ, Blackburn PR, Boczek NJ, Oliver GR, Lomberk GA, Urrutia RA, Deyle DR, and Klee EW
- Subjects
- Adult, Humans, Male, Marfan Syndrome genetics, Mutation, Mutation, Missense genetics, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Smad2 Protein genetics, Smad2 Protein metabolism, Transforming Growth Factor beta metabolism, Loeys-Dietz Syndrome genetics, Protein Serine-Threonine Kinases genetics, Receptors, Transforming Growth Factor beta genetics
- Abstract
TGF-β-related heritable connective tissue disorders are characterized by a similar pattern of cardiovascular defects, including aortic root dilatation, mitral valve prolapse, vascular aneurysms, and vascular dissections and exhibit incomplete penetrance and variable expressivity. Because of the phenotypic overlap of these disorders, panel-based genetic testing is frequently used to confirm the clinical findings. Unfortunately in many cases, variants of uncertain significance (VUSs) obscure the genetic diagnosis until more information becomes available. Here, we describe and characterize the functional impact of a novel VUS in the TGFBR2 kinase domain (c.1255G>T; p.Val419Leu), in a patient with the clinical diagnosis of Marfan syndrome spectrum. We assessed the structural and functional consequence of this VUS using molecular modeling, molecular dynamic simulations, and in vitro cell-based assays. A high-quality homology-based model of TGFBR2 was generated and computational mutagenesis followed by refinement and molecular dynamics simulations were used to assess structural and dynamic changes. Relative to wild type, the V419L induced conformational and dynamic changes that may affect ATP binding, increasing the likelihood of adopting an inactive state, and, we hypothesize, alter canonical signaling. Experimentally, we tested this by measuring the canonical TGF-β signaling pathway activation at two points; V419L significantly delayed SMAD2 phosphorylation by western blot and significantly decreased TGF-β-induced gene transcription by reporter assays consistent with known pathogenic variants in this gene. Thus, our results establish that the V419L variant leads to aberrant TGF-β signaling and confirm the diagnosis of Loeys-Dietz syndrome in this patient., (© 2017 Cousin et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2017
- Full Text
- View/download PDF
26. Novel de novo variant in EBF3 is likely to impact DNA binding in a patient with a neurodevelopmental disorder and expanded phenotypes: patient report, in silico functional assessment, and review of published cases.
- Author
-
Blackburn PR, Barnett SS, Zimmermann MT, Cousin MA, Kaiwar C, Pinto E Vairo F, Niu Z, Ferber MJ, Urrutia RA, Selcen D, Klee EW, and Pichurin PN
- Subjects
- Child, Preschool, DNA metabolism, DNA-Binding Proteins genetics, Developmental Disabilities genetics, Exome, Female, Humans, Intellectual Disability genetics, Language Development Disorders genetics, Mutation genetics, Mutation, Missense, Phenotype, Transcription Factors metabolism, Neurodevelopmental Disorders genetics, Transcription Factors genetics
- Abstract
Pathogenic variants in EBF3 were recently described in three back-to-back publications in association with a novel neurodevelopmental disorder characterized by intellectual disability, speech delay, ataxia, and facial dysmorphisms. In this report, we describe an additional patient carrying a de novo missense variant in EBF3 (c.487C>T, p.(Arg163Trp)) that falls within a conserved residue in the zinc knuckle motif of the DNA binding domain. Without a solved structure of the DNA binding domain, we generated a homology-based atomic model and performed molecular dynamics simulations for EBF3, which predicted decreased DNA affinity for p.(Arg163Trp) compared with wild-type protein and control variants. These data are in agreement with previous experimental studies of EBF1 showing the paralogous residue is essential for DNA binding. The conservation and experimental evidence existing for EBF1 and in silico modeling and dynamics simulations to validate comparable behavior of multiple variants in EBF3 demonstrates strong support for the pathogenicity of p.(Arg163Trp). We show that our patient presents with phenotypes consistent with previously reported patients harboring EBF3 variants and expands the phenotypic spectrum of this newly identified disorder with the additional feature of a bicornuate uterus.
- Published
- 2017
- Full Text
- View/download PDF
27. A novel de novo frameshift deletion in EHMT1 in a patient with Kleefstra Syndrome results in decreased H3K9 dimethylation.
- Author
-
Blackburn PR, Williams M, Cousin MA, Boczek NJ, Beek GJ, Lomberk GA, Urrutia RA, Babovic-Vuksanovic D, and Klee EW
- Abstract
Background: Kleefstra Syndrome (KS) (MIM# 610253) is an autosomal dominant disorder caused by haploinsufficiency of euchromatic histone methyltransferase-1 ( EHMT1, GLP). EHMT1 (MIM# 607001) encodes a histone methyltransferase that heterodimerizes with EHMT2 (also known as G9a, MIM# 604599), which together are responsible for mono- and dimethylation of H3 lysine 9 (H3K9me1 and -me2), resulting in transcriptional repression of target genes., Methods: This report describes an 18-year-old woman with intellectual disability, severely limited speech, hypotonia, microcephaly, and facial dysmorphisms, who was found to have a novel de novo single-base frameshift deletion in EHMT1 ., Results: Functional studies using patient fibroblasts showed decreased H3K9me2 compared to wild-type control cells, thus providing a rapid confirmatory test that complements molecular studies., Conclusion: Whole exome sequencing revealed a novel frameshift deletion in EHMT1 after a lengthy diagnostic odyssey in this patient. Functional testing using this patient's fibroblasts provides proof-of-concept for the analysis of variants of uncertain significance that are predicted to impact EHMT1 enzymatic activity.
- Published
- 2017
- Full Text
- View/download PDF
28. The Role of the Histone Methyltransferase Enhancer of Zeste Homolog 2 (EZH2) in the Pathobiological Mechanisms Underlying Inflammatory Bowel Disease (IBD).
- Author
-
Sarmento OF, Svingen PA, Xiong Y, Sun Z, Bamidele AO, Mathison AJ, Smyrk TC, Nair AA, Gonzalez MM, Sagstetter MR, Baheti S, McGovern DP, Friton JJ, Papadakis KA, Gautam G, Xavier RJ, Urrutia RA, and Faubion WA
- Subjects
- Animals, Crohn Disease pathology, Cytokines genetics, Cytokines immunology, Enhancer of Zeste Homolog 2 Protein genetics, Forkhead Transcription Factors genetics, Forkhead Transcription Factors immunology, Humans, Mice, Mice, Transgenic, T-Lymphocytes, Regulatory pathology, Th17 Cells pathology, Crohn Disease immunology, Enhancer of Zeste Homolog 2 Protein immunology, Gene Regulatory Networks immunology, T-Lymphocytes, Regulatory immunology, Th17 Cells immunology
- Abstract
Regulatory T (Treg) cells expressing the transcription factor FOXP3 play a pivotal role in maintaining immunologic self-tolerance. We and others have shown previously that EZH2 is recruited to the FOXP3 promoter and its targets in Treg cells. To further address the role for EZH2 in Treg cellular function, we have now generated mice that lack EZH2 specifically in Treg cells (EZH2
Δ/Δ FOXP3+ ). We find that EZH2 deficiency in FOXP3+ T cells results in lethal multiorgan autoimmunity. We further demonstrate that EZH2Δ/Δ FOXP3+ T cells lack a regulatory phenotype in vitro and secrete proinflammatory cytokines. Of special interest, EZH2Δ/Δ FOXP3+ mice develop spontaneous inflammatory bowel disease. Guided by these results, we assessed the FOXP3 and EZH2 gene networks by RNA sequencing in isolated intestinal CD4+ T cells from patients with Crohn's disease. Gene network analysis demonstrates that these CD4+ T cells display a Th1/Th17-like phenotype with an enrichment of gene targets shared by FOXP3 and EZH2. Combined, these results suggest that the inflammatory milieu found in Crohn's disease could lead to or result from deregulation of FOXP3/EZH2-enforced T cell gene networks contributing to the underlying intestinal inflammation., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
29. Novel Pathogenic Variant in TGFBR2 Confirmed by Molecular Modeling Is a Rare Cause of Loeys-Dietz Syndrome.
- Author
-
Zimmermann MT, Urrutia RA, Blackburn PR, Cousin MA, Boczek NJ, Klee EW, Macmurdo C, and Atwal PS
- Abstract
Loeys-Dietz syndrome (LDS) is a connective tissue disorder characterized by vascular findings of aneurysm and/or dissection of cerebral, thoracic, or abdominal arteries and skeletal findings. We report a case of a novel pathogenic variant in TGFBR2 and phenotype consistent with classic LDS. The proband was a 10-year-old presenting to the genetics clinic with an enlarged aortic root ( Z -scores 5-6), pectus excavatum, and congenital contractures of the right 2nd and 3rd digit. Molecular testing of TGFBR2 was sent to a commercial laboratory and demonstrated a novel, likely pathogenic, variant in exon 4, c.1061T>C, p.(L354P). Molecular modeling reveals alteration of local protein structure as a result of this pathogenic variant. This pathogenic variant has not been previously reported in LDS and thus expands the pathogenic variant spectrum of this condition., Competing Interests: All authors declare that there is no conflict of interests.
- Published
- 2017
- Full Text
- View/download PDF
30. Gold Nanoparticle Reprograms Pancreatic Tumor Microenvironment and Inhibits Tumor Growth.
- Author
-
Saha S, Xiong X, Chakraborty PK, Shameer K, Arvizo RR, Kudgus RA, Dwivedi SK, Hossen MN, Gillies EM, Robertson JD, Dudley JT, Urrutia RA, Postier RG, Bhattacharya R, and Mukherjee P
- Subjects
- Carcinoma, Pancreatic Ductal metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Pancreatic Neoplasms metabolism, Pancreatic Stellate Cells, Carcinoma, Pancreatic Ductal therapy, Gold, Metal Nanoparticles, Pancreatic Neoplasms therapy, Tumor Microenvironment
- Abstract
Altered tumor microenvironment (TME) arising from a bidirectional crosstalk between the pancreatic cancer cells (PCCs) and the pancreatic stellate cells (PSCs) is implicated in the dismal prognosis in pancreatic ductal adenocarcinoma (PDAC), yet effective strategies to disrupt the crosstalk is lacking. Here, we demonstrate that gold nanoparticles (AuNPs) inhibit proliferation and migration of both PCCs and PSCs by disrupting the bidirectional communication via alteration of the cell secretome. Analyzing the key proteins identified from a functional network of AuNP-altered secretome in PCCs and PSCs, we demonstrate that AuNPs impair secretions of major hub node proteins in both cell types and transform activated PSCs toward a lipid-rich quiescent phenotype. By reducing activation of PSCs, AuNPs inhibit matrix deposition, enhance angiogenesis, and inhibit tumor growth in an orthotopic co-implantation model in vivo. Auto- and heteroregulations of secretory growth factors/cytokines are disrupted by AuNPs resulting in reprogramming of the TME. By utilizing a kinase dead mutant of IRE1-α, we demonstrate that AuNPs alter the cellular secretome through the ER-stress-regulated IRE1-dependent decay pathway (RIDD) and identify endostatin and matrix metalloproteinase 9 as putative RIDD targets. Thus, AuNPs could potentially be utilized as a tool to effectively interrogate bidirectional communications in the tumor microenvironment, reprogram it, and inhibit tumor growth by its therapeutic function.
- Published
- 2016
- Full Text
- View/download PDF
31. Novel roles and mechanism for Krüppel-like factor 16 (KLF16) regulation of neurite outgrowth and ephrin receptor A5 (EphA5) expression in retinal ganglion cells.
- Author
-
Wang J, Galvao J, Beach KM, Luo W, Urrutia RA, Goldberg JL, and Otteson DC
- Published
- 2016
- Full Text
- View/download PDF
32. "The molecule's the thing:" the promise of molecular modeling and dynamic simulations in aiding the prioritization and interpretation of genomic testing results.
- Author
-
Oliver GR, Zimmermann MT, Klee EW, and Urrutia RA
- Abstract
Clinical genomics is now a reality and lies at the heart of individualized medicine efforts. The success of these approaches is evidenced by the increasing volume of publications that report causal links between genomic variants and disease. In spite of early success, clinical genomics currently faces significant challenges in establishing the relevance of the majority of variants identified by next generation sequencing tests. Indeed, the majority of mutations identified are harbored by proteins whose functions remain elusive. Herein we describe the current scenario in genomic testing and in particular the burden of variants of uncertain significance (VUSs). We highlight a role for molecular modeling and molecular dynamic simulations as tools that can significantly increase the yield of information to aid in the evaluation of pathogenicity. Though the application of these methodologies to the interpretation of variants identified by genomic testing is not yet widespread, we predict that an increase in their use will significantly benefit the mission of clinical genomics for individualized medicine.
- Published
- 2016
- Full Text
- View/download PDF
33. Krüppel-like factor KLF10 deficiency predisposes to colitis through colonic macrophage dysregulation.
- Author
-
Papadakis KA, Krempski J, Svingen P, Xiong Y, Sarmento OF, Lomberk GA, Urrutia RA, and Faubion WA
- Subjects
- Acetylation, Animals, Base Sequence, Binding Sites, CD11b Antigen metabolism, CX3C Chemokine Receptor 1, Colitis chemically induced, Colitis genetics, Colitis pathology, Colon pathology, Dextran Sulfate, Disease Models, Animal, Early Growth Response Transcription Factors genetics, Genetic Predisposition to Disease, Histocompatibility Antigens Class II metabolism, Histones metabolism, Inflammation Mediators metabolism, Interleukin-10 metabolism, Intestinal Mucosa pathology, Kruppel-Like Transcription Factors genetics, Macrophages transplantation, Mice, Knockout, Molecular Sequence Data, Phenotype, Phosphorylation, Promoter Regions, Genetic, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Receptor, Transforming Growth Factor-beta Type II, Receptors, Chemokine metabolism, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Smad2 Protein metabolism, Tumor Necrosis Factor-alpha metabolism, Colitis metabolism, Colon metabolism, Early Growth Response Transcription Factors deficiency, Intestinal Mucosa metabolism, Kruppel-Like Transcription Factors deficiency, Macrophages metabolism
- Abstract
Krüppel-like factor (KLF)-10 is an important transcriptional regulator of TGF-β1 signaling in both CD8(+) and CD4(+) T lymphocytes. In the present study, we demonstrate a novel role for KLF10 in the regulation of TGFβRII expression with functional relevance in macrophage differentiation and activation. We first show that transfer of KLF10(-/-) bone marrow-derived macrophages into wild-type (WT) mice leads to exacerbation of experimental colitis. At the cell biological level, using two phenotypic strategies, we show that KLF10-deficient mice have an altered colonic macrophage phenotype with higher frequency of proinflammatory LyC6(+)MHCII(+) cells and a reciprocal decrease of the anti-inflammatory LyC6(-)MHCII(+) subset. Additionally, the anti-inflammatory CD11b(+)CX3CR1(hi) subset of colonic macrophages is significantly decreased in KLF10(-/-) compared with WT mice under inflammatory conditions. Molecularly, CD11b(+) colonic macrophages from KLF10(-/-) mice exhibit a proinflammatory cytokine profile with increased production of TNF-α and lower production of IL-10 in response to LPS stimulation. Because KLF10 is a transcription factor, we explored how this protein may regulate macrophage function. Consequently, we analyzed the expression of TGFβRII expression in colonic macrophages and found that, in the absence of KLF10, macrophages express lower levels of TGFβRII and display an attenuated Smad-2 phosphorylation following TGF-β1 stimulation. We further show that KLF10 directly binds to the TGFβRII promoter in macrophages, leading to enhanced gene expression through histone H3 acetylation. Collectively, our data reveal a critical role for KLF10 in the epigenetic regulation of TGFβRII expression in macrophages and the acquisition of a "regulatory" phenotype that contributes to intestinal mucosal homeostasis., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
34. Biology and pathobiology of lipid droplets and their potential role in the protection of the organ of Corti.
- Author
-
Urrutia RA and Kalinec F
- Subjects
- Animals, Anti-Inflammatory Agents chemistry, Cochlea immunology, Diabetes Complications physiopathology, Ear, Inner physiology, Hearing, Hearing Loss, Sensorineural immunology, Hearing Loss, Sensorineural physiopathology, Humans, Inflammation, Obesity physiopathology, Organelles physiology, Proteomics, Signal Transduction, Cochlea physiopathology, Lipid Droplets chemistry, Organ of Corti physiology
- Abstract
The current review article seeks to extend our understanding on the role of lipid droplets within the organ of Corti. In addition to presenting an overview of the current information about the origin, structure and function of lipid droplets we draw inferences from the collective body of knowledge about this cellular organelle to build a conceptual framework to better understanding their role in auditory function. This conceptual model considers that lipid droplets play a significant role in the synthesis, storage, and release of lipids and proteins for energetic use and/or modulating cell signaling pathways. We describe the role and mechanism by which LD play a role in human diseases, and we also review emerging data from our laboratory revealing the potential role of lipid droplets from Hensen cells in the auditory organ. We suggest that lipid droplets might help to develop rapidly and efficiently the resolution phase of inflammatory responses in the mammalian cochlea, preventing inflammatory damage of the delicate inner ear structures and, consequently, sensorineural hearing loss., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Development and characterization of human-induced pluripotent stem cell-derived cholangiocytes.
- Author
-
De Assuncao TM, Sun Y, Jalan-Sakrikar N, Drinane MC, Huang BQ, Li Y, Davila JI, Wang R, O'Hara SP, Lomberk GA, Urrutia RA, Ikeda Y, and Huebert RC
- Published
- 2015
- Full Text
- View/download PDF
36. A novel role for KLF14 in T regulatory cell differentiation.
- Author
-
Sarmento OF, Svingen PA, Xiong Y, Xavier RJ, McGovern D, Smyrk TC, Papadakis KA, Urrutia RA, and Faubion WA
- Abstract
Background and Aims: KLF proteins function as epigenetic reprogramming factors during cell differentiation in many cell populations and in engineered iPS cells. In this study, we determined KLF14 function in the regulation of FOXP3, a transcription factor critical for Treg cell differentiation., Methods: We studied the effects of KLF14 on FOXP3 expression at the level of the protein and mRNA. We evaluated the functional relevance of KLF14 to FOXP3
+ Treg cells in vitro and in vivo through suppression assays and two colitis models. Finally, we analyzed the effect of KLF14 on the epigenetic landscape of the FOXP3 promoter locus through chromatin immuno-precipitation., Results: KLF14, induced upon activation of naïve CD4+ T cells, segregates to the FOXP3- population and is inversely associated with FOXP3 expression and Treg function. KLF14 KO CD4+ cells differentiated into adaptive Tregs more readily in vitro and in vivo . KLF14 KO cells demonstrated enhanced Treg suppressor function in vitro and in vivo . KLF14 repressed FOXP3 at the level of the mRNA and protein, and by ChIP assay KLF14 was found to bind to the TSDR enhancer region of FOXP3. Furthermore, loss of KLF14 reduced the levels of H3K9me3, HP1 and Suv39H1at the TSDR., Conclusions: These results outline a novel mechanism by which KLF14 regulates Treg cell differentiation via chromatin remodeling at the FOXP3 TSDR. To our knowledge, this is the first evidence supporting a role for KLF14 in maintaining the differentiated state of Treg cells and outlines a potential mechanism to modify the expression of immune genes, such as FOXP3, which are critical to T cell fate.- Published
- 2015
- Full Text
- View/download PDF
37. Krüppel-like factor KLF10 regulates transforming growth factor receptor II expression and TGF-β signaling in CD8+ T lymphocytes.
- Author
-
Papadakis KA, Krempski J, Reiter J, Svingen P, Xiong Y, Sarmento OF, Huseby A, Johnson AJ, Lomberk GA, Urrutia RA, and Faubion WA
- Subjects
- Animals, Cells, Cultured, Early Growth Response Transcription Factors deficiency, Gene Expression Regulation, Humans, Jurkat Cells, Kruppel-Like Transcription Factors deficiency, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Receptor, Transforming Growth Factor-beta Type II, CD8-Positive T-Lymphocytes metabolism, Early Growth Response Transcription Factors physiology, Kruppel-Like Transcription Factors physiology, Protein Serine-Threonine Kinases biosynthesis, Receptors, Transforming Growth Factor beta biosynthesis, Transforming Growth Factor beta biosynthesis
- Abstract
KLF10 has recently elicited significant attention as a transcriptional regulator of transforming growth factor-β1 (TGF-β1) signaling in CD4(+) T cells. In the current study, we demonstrate a novel role for KLF10 in the regulation of TGF-β receptor II (TGF-βRII) expression with functional relevance in antiviral immune response. Specifically, we show that KLF10-deficient mice have an increased number of effector/memory CD8(+) T cells, display higher levels of the T helper type 1 cell-associated transcription factor T-bet, and produce more IFN-γ following in vitro stimulation. In addition, KLF10(-/-) CD8(+) T cells show enhanced proliferation in vitro and homeostatic proliferation in vivo. Freshly isolated CD8(+) T cells from the spleen of adult mice express lower levels of surface TGF-βRII (TβRII). Congruently, in vitro activation of KLF10-deficient CD8(+) T cells upregulate TGF-βRII to a lesser extent compared with wild-type (WT) CD8(+) T cells, which results in attenuated Smad2 phosphorylation following TGF-β1 stimulation compared with WT CD8(+) T cells. Moreover, we demonstrate that KLF10 directly binds to the TGF-βRII promoter in T cells, leading to enhanced gene expression. In vivo viral infection with Daniel's strain Theiler's murine encephalomyelitis virus (TMEV) also led to lower expression of TGF-βRII among viral-specific KLF10(-/-) CD8(+) T cells and a higher percentage of IFN-γ-producing CD8(+) T cells in the spleen. Collectively, our data reveal a critical role for KLF10 in the transcriptional activation of TGF-βRII in CD8(+) T cells. Thus, KLF10 regulation of TGF-βRII in this cell subset may likely play a critical role in viral and tumor immune responses for which the integrity of the TGF-β1/TGF-βRII signaling pathway is crucial., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
38. Differential coupling of KLF10 to Sin3-HDAC and PCAF regulates the inducibility of the FOXP3 gene.
- Author
-
Xiong Y, Svingen PA, Sarmento OO, Smyrk TC, Dave M, Khanna S, Lomberk GA, Urrutia RA, and Faubion WA Jr
- Subjects
- Animals, Binding Sites, Chromatin Assembly and Disassembly, Colitis chemically induced, Colitis genetics, Colitis immunology, Colon immunology, Dextran Sulfate, Disease Models, Animal, Early Growth Response Transcription Factors chemistry, Early Growth Response Transcription Factors deficiency, Early Growth Response Transcription Factors genetics, Epigenesis, Genetic, Forkhead Transcription Factors genetics, Humans, Jurkat Cells, Kruppel-Like Transcription Factors chemistry, Kruppel-Like Transcription Factors deficiency, Kruppel-Like Transcription Factors genetics, Mice, Mice, Knockout, Models, Molecular, Mutation, Promoter Regions, Genetic, Protein Conformation, Protein Interaction Domains and Motifs, Signal Transduction, Sin3 Histone Deacetylase and Corepressor Complex chemistry, T-Lymphocytes, Regulatory immunology, Transfection, Up-Regulation, Colitis enzymology, Colon enzymology, Early Growth Response Transcription Factors metabolism, Forkhead Transcription Factors metabolism, Kruppel-Like Transcription Factors metabolism, Sin3 Histone Deacetylase and Corepressor Complex metabolism, T-Lymphocytes, Regulatory enzymology, p300-CBP Transcription Factors metabolism
- Abstract
Inducible gene expression, which requires chromatin remodeling on gene promoters, underlies the epigenetically inherited differentiation program of most immune cells. However, chromatin-mediated mechanisms that underlie these events in T regulatory cells remain to be fully characterized. Here, we report that inducibility of FOXP3, a key transcription factor for the development of T regulatory cells, depends upon Kruppel-like factor 10 (KLF10) interacting with two antagonistic histone-modifying systems. We utilized chromatin immunoprecipitation, genome-integrated reporter assays, and functional domain KLF10 mutant proteins, to characterize reciprocal interactions between this transcription factor and either the Sin3-histone deacetylase complex or the histone acetyltransferase, p300/CBP-associated factor (PCAF). We characterize a Sin3-interacting repressor domain on the NH2 terminus of KLF10, which works to limit the activating function of this transcription factor. Indeed, inactivation of this Sin3-interacting domain renders KLF10 able to physically associate with PCAF as to induce FOXP3 gene transcription. We show that this biochemical data derived from studying our genome-integrated reporter cell system are recapitulated in primary murine lymphocytes. Collectively, these results advance our understanding of how a single transcription factor, namely KLF10, functions as a toggle to integrate antagonistic signals regulating FOXP3 and, thus, immune activation., (Copyright © 2014 the American Physiological Society.)
- Published
- 2014
- Full Text
- View/download PDF
39. New role for Kruppel-like factor 14 as a transcriptional activator involved in the generation of signaling lipids.
- Author
-
de Assuncao TM, Lomberk G, Cao S, Yaqoob U, Mathison A, Simonetto DA, Huebert RC, Urrutia RA, and Shah VH
- Subjects
- Animals, Chromatin metabolism, Endothelial Cells cytology, Epigenesis, Genetic physiology, Fibroblast Growth Factor 2 metabolism, HEK293 Cells, Histones metabolism, Human Umbilical Vein Endothelial Cells, Humans, Kruppel-Like Transcription Factors genetics, Liver cytology, Lysophospholipids metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphotransferases (Alcohol Group Acceptor) genetics, Phosphotransferases (Alcohol Group Acceptor) metabolism, Sp Transcription Factors genetics, Sphingosine analogs & derivatives, Sphingosine metabolism, Transcriptional Activation physiology, Kruppel-Like Transcription Factors metabolism, Lipid Metabolism physiology, Signal Transduction physiology, Sp Transcription Factors metabolism
- Abstract
Sphingosine kinase 1 (SK1) is an FGF-inducible gene responsible for generation of sphingosine-1-phosphate, a critical lipid signaling molecule implicated in diverse endothelial cell functions. In this study, we identified SK1 as a target of the canonical FGF2/FGF receptor 1 activation pathway in endothelial cells and sought to identify novel transcriptional pathways that mediate lipid signaling. Studies using the 1.9-kb SK1 promoter and deletion mutants revealed that basal and FGF2-stimulated promoter activity occurred through two GC-rich regions located within 633 bp of the transcription start site. Screening for GC-rich binding transcription factors that could activate this site demonstrated that KLF14, a gene implicated in obesity and the metabolic syndrome, binds to this region. Congruently, overexpression of KLF14 increased basal and FGF2-stimulated SK1 promoter activity by 3-fold, and this effect was abrogated after mutation of the GC-rich sites. In addition, KLF14 siRNA transfection decreased SK1 mRNA and protein levels by 3-fold. Congruently, SK1 mRNA and protein levels were decreased in livers from KLF14 knock-out mice. Combined, luciferase, gel shift, and chromatin immunoprecipitation assays showed that KLF14 couples to p300 to increase the levels of histone marks associated with transcriptional activation (H4K8ac and H3K14ac), while decreasing repressive marks (H3K9me3 and H3K27me3). Collectively, the results demonstrate a novel mechanism whereby SK1 lipid signaling is regulated by epigenetic modifications conferred by KLF14 and p300. Thus, this is the first description of the activity and mechanisms underlying the function of KLF14 as an activator protein and novel regulator of lipid signaling., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
40. Membrane-to-nucleus signaling links insulin-like growth factor-1- and stem cell factor-activated pathways.
- Author
-
Hayashi Y, Asuzu DT, Gibbons SJ, Aarsvold KH, Bardsley MR, Lomberk GA, Mathison AJ, Kendrick ML, Shen KR, Taguchi T, Gupta A, Rubin BP, Fletcher JA, Farrugia G, Urrutia RA, and Ordog T
- Subjects
- Aminophenols pharmacology, Animals, Blotting, Western, Cell Line, Cell Line, Tumor, Cells, Cultured, Gastrointestinal Stromal Tumors genetics, Gastrointestinal Stromal Tumors metabolism, Gastrointestinal Stromal Tumors pathology, Glycogen Synthase Kinase 3 antagonists & inhibitors, Glycogen Synthase Kinase 3 metabolism, Hepatic Stellate Cells cytology, Hepatic Stellate Cells metabolism, Histones metabolism, Humans, Interstitial Cells of Cajal cytology, Interstitial Cells of Cajal metabolism, Maleimides pharmacology, Methylation drug effects, Mice, Mice, Inbred BALB C, Mice, Knockout, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit metabolism, Receptor, IGF Type 1 genetics, Receptor, IGF Type 1 metabolism, Reverse Transcriptase Polymerase Chain Reaction, Stem Cell Factor genetics, Transcription, Genetic drug effects, Cell Membrane metabolism, Cell Nucleus metabolism, Insulin-Like Growth Factor I pharmacology, Signal Transduction drug effects, Stem Cell Factor metabolism
- Abstract
Stem cell factor (mouse: Kitl, human: KITLG) and insulin-like growth factor-1 (IGF1), acting via KIT and IGF1 receptor (IGF1R), respectively, are critical for the development and integrity of several tissues. Autocrine/paracrine KITLG-KIT and IGF1-IGF1R signaling are also activated in several cancers including gastrointestinal stromal tumors (GIST), the most common sarcoma. In murine gastric muscles, IGF1 promotes Kitl-dependent development of interstitial cells of Cajal (ICC), the non-neoplastic counterpart of GIST, suggesting cooperation between these pathways. Here, we report a novel mechanism linking IGF1-IGF1R and KITLG-KIT signaling in both normal and neoplastic cells. In murine gastric muscles, the microenvironment for ICC and GIST, human hepatic stellate cells (LX-2), a model for cancer niches, and GIST cells, IGF1 stimulated Kitl/KITLG protein and mRNA expression and promoter activity by activating several signaling pathways including AKT-mediated glycogen synthase kinase-3β inhibition (GSK3i). GSK3i alone also stimulated Kitl/KITLG expression without activating mitogenic pathways. Both IGF1 and GSK3i induced chromatin-level changes favoring transcriptional activation at the Kitl promoter including increased histone H3/H4 acetylation and H3 lysine (K) 4 methylation, reduced H3K9 and H3K27 methylation and reduced occupancy by the H3K27 methyltransferase EZH2. By pharmacological or RNA interference-mediated inhibition of chromatin modifiers we demonstrated that these changes have the predicted impact on KITLG expression. KITLG knock-down and immunoneutralization inhibited the proliferation of GIST cells expressing wild-type KIT, signifying oncogenic autocrine/paracrine KITLG-KIT signaling. We conclude that membrane-to-nucleus signaling involving GSK3i establishes a previously unrecognized link between the IGF1-IGF1R and KITLG-KIT pathways, which is active in both physiologic and oncogenic contexts and can be exploited for therapeutic purposes.
- Published
- 2013
- Full Text
- View/download PDF
41. Polycomb antagonizes p300/CREB-binding protein-associated factor to silence FOXP3 in a Kruppel-like factor-dependent manner.
- Author
-
Xiong Y, Khanna S, Grzenda AL, Sarmento OF, Svingen PA, Lomberk GA, Urrutia RA, and Faubion WA Jr
- Subjects
- Animals, Chromatin Assembly and Disassembly physiology, Early Growth Response Transcription Factors genetics, Early Growth Response Transcription Factors immunology, Enhancer of Zeste Homolog 2 Protein, Forkhead Transcription Factors genetics, Forkhead Transcription Factors immunology, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors immunology, Male, Mice, Mice, Knockout, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 immunology, Polycomb Repressive Complex 2 metabolism, Polycomb-Group Proteins genetics, Polycomb-Group Proteins immunology, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory immunology, p300-CBP Transcription Factors antagonists & inhibitors, p300-CBP Transcription Factors genetics, p300-CBP Transcription Factors immunology, Early Growth Response Transcription Factors biosynthesis, Forkhead Transcription Factors biosynthesis, Gene Silencing physiology, Kruppel-Like Transcription Factors biosynthesis, Polycomb-Group Proteins metabolism, Response Elements physiology, T-Lymphocytes, Regulatory metabolism, p300-CBP Transcription Factors metabolism
- Abstract
Inducible gene expression underlies the epigenetically inherited differentiation program of most immune cells. We report that the promoter of the FOXP3 gene possesses two distinct functional states: an "off state" mediated by the polycomb histone methyltransferase complex and a histone acetyltransferase-dependent "on state." Regulating these states is the presence of a Kruppel-like factor (KLF)-containing Polycomb response element. In the KLF10(-/-) mouse, the FOXP3 promoter is epigenetically silenced by EZH2 (Enhancer of Zeste 2)-mediated trimethylation of Histone 3 K27; thus, impaired FOXP3 induction and inappropriate adaptive T regulatory cell differentiation results in vitro and in vivo. The epigenetic transmittance of adaptive T regulatory cell deficiency is demonstrated throughout more than 40 generations of mice. These results provide insight into chromatin remodeling events key to phenotypic features of distinct T cell populations.
- Published
- 2012
- Full Text
- View/download PDF
42. Inhibition of histone deacetylation potentiates the evolution of acquired temozolomide resistance linked to MGMT upregulation in glioblastoma xenografts.
- Author
-
Kitange GJ, Mladek AC, Carlson BL, Schroeder MA, Pokorny JL, Cen L, Decker PA, Wu W, Lomberk GA, Gupta SK, Urrutia RA, and Sarkaria JN
- Subjects
- Acetylation drug effects, Animals, Antineoplastic Agents, Alkylating pharmacology, Blotting, Western, DNA Methylation drug effects, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Dacarbazine pharmacology, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma metabolism, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids pharmacology, Kaplan-Meier Estimate, Methylation drug effects, Mice, Promoter Regions, Genetic genetics, Reverse Transcriptase Polymerase Chain Reaction, Temozolomide, Tumor Suppressor Proteins genetics, Up-Regulation drug effects, Vorinostat, Xenograft Model Antitumor Assays, DNA Modification Methylases metabolism, DNA Repair Enzymes metabolism, Dacarbazine analogs & derivatives, Glioblastoma drug therapy, Histones metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Purpose: The therapeutic benefit of temozolomide in glioblastoma multiforme (GBM) is limited by resistance. The goal of this study was to elucidate mechanisms of temozolomide resistance in GBM., Experimental Design: We developed an in vivo GBM model of temozolomide resistance and used paired parental and temozolomide-resistant tumors to define the mechanisms underlying the development of resistance and the influence of histone deacetylation (HDAC) inhibition., Results: Analysis of paired parental and resistant lines showed upregulation of O6-methylguanine-DNA methyltransferase (MGMT) expression in 3 of the 5 resistant xenografts. While no significant change was detected in MGMT promoter methylation between parental and derivative-resistant samples, chromatin immunoprecipitation showed an association between MGMT upregulation and elevated acetylation of lysine 9 of histone H3 (H3K9-ac) and decreased dimethylation (H3K9-me2) in GBM12 and GBM14. In contrast, temozolomide resistance development in GBM22 was not linked to MGMT expression, and both parental and resistant lines had low H3K9-ac and high H3K9-me2 within the MGMT promoter. In the GBM12TMZ-resistant line, MGMT reexpression was accompanied by increased recruitment of SP1, C-JUN, NF-κB, and p300 within the MGMT promoter. Interestingly, combined treatment of GBM12 flank xenografts with temozolomide and the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) favored the evolution of temozolomide resistance by MGMT overexpression as compared with treatment with temozolomide alone., Conclusion: This study shows, for the first time, a unique mechanism of temozolomide resistance development driven by chromatin-mediated MGMT upregulation and highlights the potential for epigenetically directed therapies to influence the mechanisms of resistance development in GBM.
- Published
- 2012
- Full Text
- View/download PDF
43. Immortalized liver endothelial cells: a cell culture model for studies of motility and angiogenesis.
- Author
-
Huebert RC, Jagavelu K, Liebl AF, Huang BQ, Splinter PL, LaRusso NF, Urrutia RA, and Shah VH
- Subjects
- Animals, Cell Culture Techniques methods, Cell Transformation, Viral immunology, Chemotaxis immunology, Endothelial Cells ultrastructure, Immunomagnetic Separation, Lentivirus immunology, Liver Diseases immunology, Mice, Platelet Endothelial Cell Adhesion Molecule-1 immunology, von Willebrand Factor immunology, Cell Line, Transformed cytology, Cell Movement immunology, Endothelial Cells cytology, Endothelial Cells immunology, Liver cytology, Liver immunology, Neovascularization, Physiologic immunology
- Abstract
Hepatic sinusoidal endothelial cells (HSECs) are a unique subpopulation of fenestrated endothelial cells lining the hepatic sinusoids and comprising the majority of endothelial cells within the liver. HSECs not only have important roles in blood clearance, vascular tone, and immunity, but also undergo pathological changes, contributing to fibrosis, angiogenesis, and portal hypertension. There are few cell culture models for in vitro studies of motility and angiogenesis as primary cells are time-consuming to isolate, are limited in number, and often lack features of pathological vasculature. The aim of this study was to generate an immortalized cell line derived from HSECs that mimic pathological vasculature and allows detailed molecular interventions to be pursued. HSECs were isolated from mouse liver using CD31-based immunomagnetic separation, immortalized with SV40 large T-antigen, and subcloned on the basis of their ability to endocytose the acetylated low-density lipoprotein (AcLDL). The resulting cell line, transformed sinusoidal endothelial cells (TSECs), maintains an endothelial phenotype as well as some HSEC-specific features. This is evidenced by typical microscopic features of endothelia, including formation of lamellipodia and filopodia, and a cobblestone morphology of cell monolayers. Electron microscopy showed maintenance of a limited number of fenestrae organized in sieve plates. TSECs express numerous endothelia-specific markers, including CD31 and von Willebrand's factor (vWF), as detected by PCR array, immunoblotting, and immunofluorescence (IF). Functionally, TSECs maintain a number of key endothelial features, including migration in response to angiogenic factors, formation of vascular tubes, endocytosis of AcLDL, and remodeling of extracellular matrix. Their phenotype most closely resembles the pathological neovasculature associated with chronic liver disease, in which cells become proliferative, defenestrated, and angiogenic. Importantly, the cells can be transduced efficiently with viral vectors. TSECs should provide a reproducible cell culture model for high-throughput in vitro studies pertaining to a broad range of liver endothelial cell functions, but likely broader endothelial cell biology as well.
- Published
- 2010
- Full Text
- View/download PDF
44. Neuropilin-1 mediates divergent R-Smad signaling and the myofibroblast phenotype.
- Author
-
Cao Y, Szabolcs A, Dutta SK, Yaqoob U, Jagavelu K, Wang L, Leof EB, Urrutia RA, Shah VH, and Mukhopadhyay D
- Subjects
- Actins genetics, Actins metabolism, Animals, Cell Line, Transformed, Fibroblasts cytology, Gene Expression Regulation physiology, Hepatic Stellate Cells cytology, Hepatic Stellate Cells metabolism, Humans, Liver metabolism, Mice, Mice, Knockout, Myoblasts cytology, Neuropilin-1 genetics, Phosphorylation physiology, Smad Proteins, Receptor-Regulated genetics, Transforming Growth Factor beta1 metabolism, Fibroblasts metabolism, Myoblasts metabolism, Neuropilin-1 metabolism, Signal Transduction physiology, Smad Proteins, Receptor-Regulated metabolism
- Abstract
The transforming growth factor-beta (TGF-β) superfamily is one of the most diversified cell signaling pathways and regulates many physiological and pathological processes. Recently, neuropilin-1 (NRP-1) was reported to bind and activate the latent form of TGF-β1 (LAP-TGF-β1). We investigated the role of NRP-1 on Smad signaling in stromal fibroblasts upon TGF-β stimulation. Elimination of NRP-1 in stromal fibroblast cell lines increases Smad1/5 phosphorylation and downstream responses as evidenced by up-regulation of inhibitor of differentiation (Id-1). Conversely, NRP-1 loss decreases Smad2/3 phosphorylation and its responses as shown by down-regulation of α-smooth muscle actin (α-SMA) and also cells exhibit more quiescent phenotypes and growth arrest. Moreover, we also observed that NRP-1 expression is increased during the culture activation of hepatic stellate cells (HSCs), a liver resident fibroblast. Taken together, our data suggest that NRP-1 functions as a key determinant of the diverse responses downstream of TGF-β1 that are mediated by distinct Smad proteins and promotes myofibroblast phenotype.
- Published
- 2010
- Full Text
- View/download PDF
45. Kitlow stem cells cause resistance to Kit/platelet-derived growth factor alpha inhibitors in murine gastrointestinal stromal tumors.
- Author
-
Bardsley MR, Horváth VJ, Asuzu DT, Lorincz A, Redelman D, Hayashi Y, Popko LN, Young DL, Lomberk GA, Urrutia RA, Farrugia G, Rubin BP, and Ordog T
- Subjects
- Animals, Antigens, CD34 analysis, Benzamides, Biomarkers, Tumor metabolism, Cell Differentiation, Cell Proliferation, Cell Survival, Cells, Cultured, Clone Cells, Dose-Response Relationship, Drug, Down-Regulation, Gastrointestinal Stromal Tumors genetics, Gastrointestinal Stromal Tumors metabolism, Hyaluronan Receptors analysis, Hyperplasia, Imatinib Mesylate, Interstitial Cells of Cajal immunology, Interstitial Cells of Cajal metabolism, Interstitial Cells of Cajal pathology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Nude, Mutation, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Piperazines pharmacology, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit metabolism, Pyrans pharmacology, Pyrimidines pharmacology, Receptor, Platelet-Derived Growth Factor alpha metabolism, Time Factors, Tumor Burden, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Gastrointestinal Stromal Tumors pathology, Interstitial Cells of Cajal drug effects, Neoplastic Stem Cells drug effects, Proto-Oncogene Proteins c-kit antagonists & inhibitors, Receptor, Platelet-Derived Growth Factor alpha antagonists & inhibitors
- Abstract
Background & Aims: Gastrointestinal stromal tumors (GIST) are related to interstitial cells of Cajal (ICC) and often contain activating stem cell factor receptor (Kit) or platelet-derived growth factor receptor alpha (Pdgfra) mutations. Kit/Pdgfra inhibitors such as imatinib mesylate have increased progression-free survival in metastatic GIST but are not curative. In mouse models we investigated whether Kit(low) ICC progenitors could represent an inherently Kit/Pdgfra inhibitor-resistant reservoir for GIST., Methods: Isolated Kit(low)Cd44(+)Cd34(+) cells were characterized after serial cloning. The tumorigenic potential of spontaneously transformed cells was investigated in nude mice. The Kit(low)Cd44(+)Cd34(+) cells' responsiveness to Kit activation and blockade was studied by enumerating them in Kit(K641E) mice (a GIST model), in mice with defective Kit signaling, and pharmacologically., Results: Single isolated Kit(low)Cd44(+)Cd34(+) cells were clonogenic and capable of self-renewal and differentiation into ICC. In nude mice, spontaneously transformed cells formed malignant tumors expressing GIST markers. The Kit(low)Cd44(+)Cd34(+) cells were resistant to in vitro Kit blockade, including by imatinib, and occurred in normal numbers in mice with reduced Kit signaling. In Kit(K641E) mice, the mutant ICC stem cells were grossly hyperplastic but remained imatinib-resistant. In contrast, the cancer stem, cell-targeting drug salinomycin blocked the proliferation of Kit(low)Cd44(+)Cd34(+) cells and increased their sensitivity to imatinib., Conclusions: Kit(low)Cd44(+)Cd34(+) progenitors are true stem cells for normal and hyperplastic ICC and give rise to GIST. Resistance to Kit/Pdgfra inhibitors is inherent in GIST and is caused by the native ICC stem cells' lack of dependence on Kit for survival, which is maintained after the acquisition of oncogenic Kit mutation. Cancer stem cell drugs may target these cells., (Copyright © 2010 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. 2007 American Pancreatic Association Presidential Address: a proud member of the APA.
- Author
-
Urrutia RA
- Subjects
- Biomedical Research, Humans, Research Support as Topic, Pancreas, Pancreatic Diseases, Societies, Medical economics, Societies, Medical organization & administration, Societies, Medical trends
- Published
- 2008
- Full Text
- View/download PDF
47. A screen of candidate genes and influence of beta2-adrenergic receptor genotypes in postural tachycardia syndrome.
- Author
-
Nickander KK, Carlson PJ, Urrutia RA, Camilleri M, and Low PA
- Subjects
- Adult, Blood Pressure genetics, Electron Transport Complex I genetics, Female, Genotype, Heart Rate genetics, Humans, Male, Middle Aged, Norepinephrine Plasma Membrane Transport Proteins, Posture, Symporters genetics, Genetic Testing, Polymorphism, Single Nucleotide, Receptors, Adrenergic, beta-2 genetics, Tachycardia genetics
- Abstract
Objective: To screen candidate genes, encoding beta2-adrenergic receptor (beta2AR), alpha2C-adrenergic receptor (alpha(2C)AR), norepinephrine transporter (NET), and mitochondrial complex I (COI), for common single nucleotide polymorphisms (SNPs) in patients with postural tachycardia syndrome (POTS); alterations could potentially cause or aggravate orthostatic tachycardia and to relate beta2AR SNPs, known to effect venomotor tone, to heart rate (HR) and blood pressure measurements during 10-min head-up tilt., Methods: (a) DNA extraction from leukocytes of 29 patients with POTS; (b) Denaturing high performance liquid chromatography analysis to screen for the 12-bp deletion (Del322-325) in alpha(2C)AR and for the alanine to proline mutation at amino acid 457 (Ala457Pro) in NET; (c) Systematic direct sequence analysis to screen for SNPs in beta2AR, NET, and COI., Results: Three common polymorphisms were abundant in at least one allele in beta2AR resulting in a cysteine to arginine in the 5' promoter region (72% of patients), an arginine to glycine at amino acid-16 (Gly16; 86%), and a glutamine to glutamic acid at amino acid-27 (Glu27; 66%), a frequency that was no different to the normal Caucasian population. Orthostatic HR was significantly greater in patients with Glu27. Diastolic blood pressure (DBP) was significantly lower in a subset of patients with Gly16 whose HR were > or =120 beats/min with head-up tilt. All patients did not show the Ala457Pro mutation of NET; all sequence variants detected in alpha(2C)AR, NET, and COI were not considered causally related to POTS., Conclusions: Of the candidate genes screened, none harbored a SNP considered to be causally related to POTS. There was significant association of HR and DBP with SNPs of the gene encoding beta2AR; Gly16 or Glu27 could aggravate orthostatic tachycardia by excessive venous pooling.
- Published
- 2005
- Full Text
- View/download PDF
48. Glycogen synthase kinase-3beta participates in nuclear factor kappaB-mediated gene transcription and cell survival in pancreatic cancer cells.
- Author
-
Ougolkov AV, Fernandez-Zapico ME, Savoy DN, Urrutia RA, and Billadeau DD
- Subjects
- Apoptosis drug effects, Apoptosis physiology, Cell Growth Processes physiology, Cell Line, Tumor, Cell Survival physiology, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic physiology, Glycogen Synthase Kinase 3 antagonists & inhibitors, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Pancreatic Neoplasms pathology, Thiazoles pharmacology, Urea pharmacology, Glycogen Synthase Kinase 3 physiology, NF-kappa B physiology, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms genetics, Transcription, Genetic physiology, Urea analogs & derivatives
- Abstract
Recent studies using glycogen synthase kinase-3beta (GSK-3beta)-deficient mouse embryonic fibroblasts suggest that GSK-3beta positively regulates nuclear factor kappaB (NFkappaB)-mediated gene transcription. Because NFkappaB is suggested to participate in cell proliferation and survival pathways in pancreatic cancer, we investigated the role of GSK-3beta in regulating these cellular processes. Herein, we show that pancreatic cancer cells contain a pool of active GSK-3beta and that pharmacologic inhibition of GSK-3 kinase activity using small molecule inhibitors or genetic depletion of GSK-3beta by RNA interference leads to decreased cancer cell proliferation and survival. Mechanistically, we show that GSK-3beta influences NFkappaB-mediated gene transcription at a point distal to the Ikappa kinase complex, as only ectopic expression of the NFkappaB subunits p65/p50, but not an Ikappa kinase beta constitutively active mutant, could rescue the decreased cellular proliferation and survival associated with GSK-3beta inhibition. Taken together, our results simultaneously identify a previously unrecognized role for GSK-3beta in cancer cell survival and proliferation and suggest GSK-3beta as a potential therapeutic target in the treatment of pancreatic cancer.
- Published
- 2005
- Full Text
- View/download PDF
49. Upregulation of dynamin II expression during the acquisition of a mature pancreatic acinar cell phenotype.
- Author
-
Cook TA, Mesa KJ, Gebelein BA, and Urrutia RA
- Subjects
- Animals, Cell Differentiation drug effects, Cell Line, Dexamethasone pharmacology, Dynamins, GTP Phosphohydrolases metabolism, Pancreas cytology, Phenotype, RNA, Messenger genetics, Rats, GTP Phosphohydrolases genetics, Pancreas immunology, Up-Regulation
- Abstract
Members of the dynamin superfamily are GTPases which have been shown to support receptor-mediated endocytosis in vivo and bind to growth factor receptor-associated proteins in vitro. In acinar cells of the pancreas, receptor-mediated endocytosis is very important for the recycling of membranes after secretory granule release. Therefore, characterization of the molecular machinery responsible for this process is critical for a better understanding of this phenomenon. In this study we sought to determine the expression pattern of the endocytic GTPase dynamin II during pancreatic acinar cell differentiation in developing rat embryos and in dexamethasone-treated AR42J cells using Western blot, Northern blot, and immunocytochemical analyses. During pancreatic development, dynamin immunoreactivity is almost undetectable until day E17 but undergoes significant upregulation in acinar cells starting at E18. In addition, the levels of dynamin mRNA and protein in AR42J cells increase approximately threefold during dexamethasone-induced acinar differentiation. The increase in dynamin levels that occurs in both embryonic pancreatic cells and dexamethasone-treated AR42J cells correlates with the establishment of a more differentiated acinar phenotype. Therefore, these results suggest a potential role for dynamin in supporting receptor-mediated endocytosis in mature pancreatic acinar cells.
- Published
- 1996
- Full Text
- View/download PDF
50. The Dictyostelium myosin IE heavy chain gene encodes a truncated isoform that lacks sequences corresponding to the actin binding site in the tail.
- Author
-
Urrutia RA, Jung G, and Hammer JA 3rd
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Binding Sites, Cloning, Molecular, Gene Expression, Molecular Sequence Data, Myosins chemistry, RNA, Messenger biosynthesis, Dictyostelium genetics, Myosins genetics
- Abstract
We have isolated cDNA and genomic clones which together span the entire coding sequence for the 114.8 kDa heavy chain of Dictyostelium myosin IE (DMIE). The deduced primary sequence reveals a pattern characteristic of all myosins I, i.e., a myosin-like globular head domain fused to a tail domain that shows no similarity to the coiled-coil rod-like tail of type II myosins. The approx. 35 kDa tail domain of DMIE shows some sequence similarity to the membrane interaction region of other myosins I (tail-homology-region 1; TH-1), but lacks completely the sequences that correspond to the second actin binding site (the glycine-, proline- and alanine-rich TH-2 region and the src-like TH-3 region). Therefore, DMIE more closely resembles DMIA (Titus et al. (1989) Cell Regul 1, 55-63), which is also truncated, than DMIB and DMID, both of which possess all three tail homology regions. The similarity between the DMIE and DMIA isoforms extends to their pattern of expression, in which the steady state level of transcript for both genes is highest in vegetative cells and falls gradually after five to ten hours of starvation-induced development. Together, these results have important implications for interpreting and prioritizing gene targeting experiments designed to identify the functions of myosins I in vivo.
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.