Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
Cell ; 162(4): 766-79, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276631

ABSTRACT

Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.


Subject(s)
Hepatocytes/transplantation , Liver/cytology , Liver/physiology , Animals , Bile Ducts/cytology , Cell Proliferation , Cell Transplantation/methods , Hepatocytes/classification , Hepatocytes/cytology , Liver/injuries , Liver Neoplasms , Mice , Regeneration , SOX9 Transcription Factor/genetics , Transcriptome
2.
Mol Cell ; 70(5): 842-853.e7, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861157

ABSTRACT

Heterochromatic repetitive satellite RNAs are extensively transcribed in a variety of human cancers, including BRCA1 mutant breast cancer. Aberrant expression of satellite RNAs in cultured cells induces the DNA damage response, activates cell cycle checkpoints, and causes defects in chromosome segregation. However, the mechanism by which satellite RNA expression leads to genomic instability is not well understood. Here we provide evidence that increased levels of satellite RNAs in mammary glands induce tumor formation in mice. Using mass spectrometry, we further show that genomic instability induced by satellite RNAs occurs through interactions with BRCA1-associated protein networks required for the stabilization of DNA replication forks. Additionally, de-stabilized replication forks likely promote the formation of RNA-DNA hybrids in cells expressing satellite RNAs. These studies lay the foundation for developing novel therapeutic strategies that block the effects of non-coding satellite RNAs in cancer cells.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , DNA Damage , Genomic Instability , Heterochromatin/genetics , RNA, Neoplasm/genetics , RNA, Satellite/genetics , Animals , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Heterochromatin/metabolism , Humans , MCF-7 Cells , Mice , Protein Binding , RNA, Neoplasm/metabolism , RNA, Satellite/metabolism , Tumor Burden
3.
Nature ; 553(7688): 351-355, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29320480

ABSTRACT

The circadian clock imposes daily rhythms in cell proliferation, metabolism, inflammation and DNA damage response. Perturbations of these processes are hallmarks of cancer and chronic circadian rhythm disruption predisposes individuals to tumour development. This raises the hypothesis that pharmacological modulation of the circadian machinery may be an effective therapeutic strategy for combating cancer. REV-ERBs, the nuclear hormone receptors REV-ERBα (also known as NR1D1) and REV-ERBß (also known as NR1D2), are essential components of the circadian clock. Here we show that two agonists of REV-ERBs-SR9009 and SR9011-are specifically lethal to cancer cells and oncogene-induced senescent cells, including melanocytic naevi, and have no effect on the viability of normal cells or tissues. The anticancer activity of SR9009 and SR9011 affects a number of oncogenic drivers (such as HRAS, BRAF, PIK3CA and others) and persists in the absence of p53 and under hypoxic conditions. The regulation of autophagy and de novo lipogenesis by SR9009 and SR9011 has a critical role in evoking an apoptotic response in malignant cells. Notably, the selective anticancer properties of these REV-ERB agonists impair glioblastoma growth in vivo and improve survival without causing overt toxicity in mice. These results indicate that pharmacological modulation of circadian regulators is an effective antitumour strategy, identifying a class of anticancer agents with a wide therapeutic window. We propose that REV-ERB agonists are inhibitors of autophagy and de novo lipogenesis, with selective activity towards malignant and benign neoplasms.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Oncogenes/genetics , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Circadian Clocks/genetics , Circadian Clocks/physiology , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Lipogenesis/drug effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Neoplasms/genetics , Nevus/drug therapy , Nevus/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Thiophenes/pharmacology
4.
Cell ; 134(3): 485-95, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18692471

ABSTRACT

Mast cells are known to play a pivotal role in allergic diseases. Cross-linking of the high-affinity receptor for IgE (FcepsilonRI) leads to degranulation and allergic inflammation; however, the regulatory mechanisms of IgE-dependent exocytosis remain unknown. We show here that IkappaB kinase (IKK) 2 in mast cells plays critical roles in IgE-mediated anaphylaxis in vivo, and IgE-mediated degranulation in vitro, in an NF-kB-independent manner. Upon FcvarepsilonRI stimulation, IKK2 phosphorylates SNAP-23, the target membrane soluble N-ethylmaleimide-sensitive fusion factor attachment protein receptor (SNARE), and ectopic expression of a phospho-mimetic mutant of SNAP-23 partially rescued the impaired IgE-mediated degranulation in IKK2-deficient mast cells. These results suggest that IKK2 phosphorylation of SNAP-23 leads to degranulation and anaphylactic reactions. While this reaction is NF-kB-independent, we additionally show that IKK2 also regulates late-phase allergic reactions promoted by the release of proinflammatory cytokines in an NF-kB-dependent manner. The findings suggest that IKK2 is a central player in allergic reactions.


Subject(s)
Cell Degranulation , I-kappa B Kinase/metabolism , Mast Cells/cytology , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Anaphylaxis/immunology , Animals , Immunoglobulin E/immunology , Mast Cells/immunology , Mice , Phosphorylation
5.
Proc Natl Acad Sci U S A ; 116(15): 7363-7370, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30918126

ABSTRACT

Coordination of growth and genomic stability is critical for normal cell physiology. Although the E3 ubiquitin ligase BRCA1 is a key player in maintenance of genomic stability, its role in growth signaling remains elusive. Here, we show that BRCA1 facilitates stabilization of YAP1 protein and turning "off" the Hippo pathway through ubiquitination of NF2. In BRCA1-deficient cells Hippo pathway is "turned On." Phosphorylation of YAP1 is crucial for this signaling process because a YAP1 mutant harboring alanine substitutions (Mt-YAP5SA) in LATS1 kinase recognition sites not only resists degradation but also rescues YAP1 transcriptional activity in BRCA1-deficient cells. Furthermore, an ectopic expression of the active Mt-YAP5SA, but not inactive Mt-YAP6SA, promotes EGF-independent proliferation and tumorigenesis in BRCA1-/- mammary epithelial cells. These findings establish an important role of BRCA1 in regulating stability of YAP1 protein that correlates positively with cell proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , HEK293 Cells , Hippo Signaling Pathway , Humans , Mutation, Missense , Neurofibromin 2/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , YAP-Signaling Proteins
7.
Proc Natl Acad Sci U S A ; 115(11): 2557-2560, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29487213

ABSTRACT

In keeping with the growing movement in scientific publishing toward transparency in data and methods, we propose changes to journal authorship policies and procedures to provide insight into which author is responsible for which contributions, better assurance that the list is complete, and clearly articulated standards to justify earning authorship credit. To accomplish these goals, we recommend that journals adopt common and transparent standards for authorship, outline responsibilities for corresponding authors, adopt the Contributor Roles Taxonomy (CRediT) (docs.casrai.org/CRediT) methodology for attributing contributions, include this information in article metadata, and require authors to use the ORCID persistent digital identifier (https://orcid.org). Additionally, we recommend that universities and research institutions articulate expectations about author roles and responsibilities to provide a point of common understanding for discussion of authorship across research teams. Furthermore, we propose that funding agencies adopt the ORCID identifier and accept the CRediT taxonomy. We encourage scientific societies to further authorship transparency by signing on to these recommendations and promoting them through their meetings and publications programs.

8.
Mol Cell ; 48(5): 771-84, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23123196

ABSTRACT

Many types of human tumor cells have overexpressed pyruvate kinase M2 (PKM2). However, the mechanism underlying this increased PKM2 expression remains to be defined. We demonstrate here that EGFR activation induces PLCγ1-dependent PKCε monoubiquitylation at Lys321 mediated by RINCK1 ubiquitin ligase. Monoubiquitylated PKCε interacts with a ubiquitin-binding domain in NEMO zinc finger and recruits the cytosolic IKK complex to the plasma membrane, where PKCε phosphorylates IKKß at Ser177 and activates IKKß. Activated RelA interacts with HIF1α, which is required for RelA to bind the PKM promoter. PKCε- and NF-κB-dependent PKM2 upregulation is required for EGFR-promoted glycolysis and tumorigenesis. In addition, PKM2 expression correlates with EGFR and IKKß activity in human glioblastoma specimens and with grade of glioma malignancy. These findings highlight the distinct regulation of NF-κB by EGF, in contrast to TNF-α, and the importance of the metabolic cooperation between the EGFR and NF-κB pathways in PKM2 upregulation and tumorigenesis.


Subject(s)
Brain Neoplasms/enzymology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Glioblastoma/enzymology , Membrane Proteins/metabolism , NF-kappa B/metabolism , Protein Kinase C-epsilon/metabolism , Thyroid Hormones/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glioblastoma/genetics , Glioblastoma/pathology , Glucose/metabolism , Glycolysis , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , I-kappa B Kinase/metabolism , Lactic Acid/metabolism , Membrane Proteins/genetics , Mice , Mice, Nude , Mutagenesis, Site-Directed , Mutation , NF-kappa B/genetics , Neoplasm Grading , Neoplasm Transplantation , Phospholipase C gamma/metabolism , Phosphorylation , Polypyrimidine Tract-Binding Protein/metabolism , Prognosis , Promoter Regions, Genetic , Protein Kinase C-epsilon/genetics , RNA Interference , Serine , Signal Transduction , Thyroid Hormones/genetics , Transcription Factor RelA/metabolism , Transfection , Ubiquitination , Up-Regulation , Thyroid Hormone-Binding Proteins
9.
Proc Natl Acad Sci U S A ; 114(10): E1941-E1950, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28202722

ABSTRACT

Safe and efficient delivery of messenger RNAs for protein replacement therapies offers great promise but remains challenging. In this report, we demonstrate systemic, in vivo, nonviral mRNA delivery through lipid nanoparticles (LNPs) to treat a Factor IX (FIX)-deficient mouse model of hemophilia B. Delivery of human FIX (hFIX) mRNA encapsulated in our LUNAR LNPs results in a rapid pulse of FIX protein (within 4-6 h) that remains stable for up to 4-6 d and is therapeutically effective, like the recombinant human factor IX protein (rhFIX) that is the current standard of care. Extensive cytokine and liver enzyme profiling showed that repeated administration of the mRNA-LUNAR complex does not cause any adverse innate or adaptive immune responses in immune-competent, hemophilic mice. The levels of hFIX protein that were produced also remained consistent during repeated administrations. These results suggest that delivery of long mRNAs is a viable therapeutic alternative for many clotting disorders and for other hepatic diseases where recombinant proteins may be unaffordable or unsuitable.


Subject(s)
Drug Carriers/administration & dosage , Factor IX/pharmacokinetics , Hemophilia B/therapy , Nanoparticles/administration & dosage , RNA, Messenger/pharmacokinetics , Animals , Cholesterol/chemistry , Cytokines/metabolism , Disease Models, Animal , Drug Compounding/methods , Drug Evaluation, Preclinical , Factor IX/genetics , Factor IX/metabolism , Female , Genetic Therapy/methods , Hemophilia B/genetics , Hemophilia B/metabolism , Hemophilia B/pathology , Humans , Hydrogen-Ion Concentration , Injections, Intravenous , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Phosphatidylcholines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics
10.
Genes Dev ; 26(24): 2684-9, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23249732

ABSTRACT

Centrioles organize the centrosome, and accurate control of their number is critical for the maintenance of genomic integrity. Centriole duplication occurs once per cell cycle and is controlled by Polo-like kinase 4 (Plk4). We showed previously that Plk4 phosphorylates itself to promote its degradation by the proteasome. Here we demonstrate that this autoregulated instability controls the abundance of endogenous Plk4. Preventing Plk4 autoregulation causes centrosome amplification, stabilization of p53, and loss of cell proliferation; moreover, suppression of p53 allows growth of cells carrying amplified centrosomes. Plk4 autoregulation thus guards against genome instability by limiting centrosome duplication to once per cell cycle.


Subject(s)
Cell Cycle/physiology , Centrosome/physiology , Protein Serine-Threonine Kinases/metabolism , Cell Division/genetics , Cell Line , Cell Proliferation , Enzyme Stability/physiology , Gene Targeting , Homeostasis/physiology , Humans , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/metabolism
11.
BMC Bioinformatics ; 20(1): 294, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142274

ABSTRACT

BACKGROUND: Biochemical networks are often described through static or time-averaged measurements of the component macromolecules. Temporal variation in these components plays an important role in both describing the dynamical nature of the network as well as providing insights into causal mechanisms. Few methods exist, specifically for systems with many variables, for analyzing time series data to identify distinct temporal regimes and the corresponding time-varying causal networks and mechanisms. RESULTS: In this study, we use well-constructed temporal transcriptional measurements in a mammalian cell during a cell cycle, to identify dynamical networks and mechanisms describing the cell cycle. The methods we have used and developed in part deal with Granger causality, Vector Autoregression, Estimation Stability with Cross Validation and a nonparametric change point detection algorithm that enable estimating temporally evolving directed networks that provide a comprehensive picture of the crosstalk among different molecular components. We applied our approach to RNA-seq time-course data spanning nearly two cell cycles from Mouse Embryonic Fibroblast (MEF) primary cells. The change-point detection algorithm is able to extract precise information on the duration and timing of cell cycle phases. Using Least Absolute Shrinkage and Selection Operator (LASSO) and Estimation Stability with Cross Validation (ES-CV), we were able to, without any prior biological knowledge, extract information on the phase-specific causal interaction of cell cycle genes, as well as temporal interdependencies of biological mechanisms through a complete cell cycle. CONCLUSIONS: The temporal dependence of cellular components we provide in our model goes beyond what is known in the literature. Furthermore, our inference of dynamic interplay of multiple intracellular mechanisms and their temporal dependence on one another can be used to predict time-varying cellular responses, and provide insight on the design of precise experiments for modulating the regulation of the cell cycle.


Subject(s)
Cell Cycle/genetics , Gene Regulatory Networks , Algorithms , Animals , Cell Cycle Checkpoints/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , G1 Phase/genetics , Genes, cdc , Mice , Time Factors
12.
Proc Natl Acad Sci U S A ; 112(47): E6476-85, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26542681

ABSTRACT

Lung adenocarcinoma, a major form of non-small cell lung cancer, is the leading cause of cancer deaths. The Cancer Genome Atlas analysis of lung adenocarcinoma has identified a large number of previously unknown copy number alterations and mutations, requiring experimental validation before use in therapeutics. Here, we describe an shRNA-mediated high-throughput approach to test a set of genes for their ability to function as tumor suppressors in the background of mutant KRas and WT Tp53. We identified several candidate genes from tumors originated from lentiviral delivery of shRNAs along with Cre recombinase into lungs of Loxp-stop-Loxp-KRas mice. Ephrin receptorA2 (EphA2) is among the top candidate genes and was reconfirmed by two distinct shRNAs. By generating knockdown, inducible knockdown and knockout cell lines for loss of EphA2, we showed that negating its expression activates a transcriptional program for cell proliferation. Loss of EPHA2 releases feedback inhibition of KRAS, resulting in activation of ERK1/2 MAP kinase signaling, leading to enhanced cell proliferation. Intriguingly, loss of EPHA2 induces activation of GLI1 transcription factor and hedgehog signaling that further contributes to cell proliferation. Small molecules targeting MEK1/2 and Smoothened hamper proliferation in EphA2-deficient cells. Additionally, in EphA2 WT cells, activation of EPHA2 by its ligand, EFNA1, affects KRAS-RAF interaction, leading to inhibition of the RAS-RAF-MEK-ERK pathway and cell proliferation. Together, our studies have identified that (i) EphA2 acts as a KRas cooperative tumor suppressor by in vivo screen and (ii) reactivation of the EphA2 signal may serve as a potential therapeutic for KRas-induced human lung cancers.


Subject(s)
Adenocarcinoma/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, EphA2/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma of Lung , Animals , Base Sequence , Carcinogenesis/pathology , Cell Proliferation , Enzyme Activation , Gene Knockdown Techniques , Genome, Human , Hedgehog Proteins/metabolism , High-Throughput Screening Assays , Humans , Ligands , MAP Kinase Signaling System , Mice, Knockout , Molecular Sequence Data , Mutation/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
13.
Nature ; 477(7363): 179-84, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21901007

ABSTRACT

Mutations in the tumour suppressor gene BRCA1 lead to breast and/or ovarian cancer. Here we show that loss of Brca1 in mice results in transcriptional de-repression of the tandemly repeated satellite DNA. Brca1 deficiency is accompanied by a reduction of condensed DNA regions in the genome and loss of ubiquitylation of histone H2A at satellite repeats. BRCA1 binds to satellite DNA regions and ubiquitylates H2A in vivo. Ectopic expression of H2A fused to ubiquitin reverses the effects of BRCA1 loss, indicating that BRCA1 maintains heterochromatin structure via ubiquitylation of histone H2A. Satellite DNA de-repression was also observed in mouse and human BRCA1-deficient breast cancers. Ectopic expression of satellite DNA can phenocopy BRCA1 loss in centrosome amplification, cell-cycle checkpoint defects, DNA damage and genomic instability. We propose that the role of BRCA1 in maintaining global heterochromatin integrity accounts for many of its tumour suppressor functions.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/genetics , Gene Silencing , Genes, BRCA1/physiology , Heterochromatin/genetics , Heterochromatin/metabolism , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Breast/cytology , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , DNA, Satellite/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , HeLa Cells , Histones/metabolism , Humans , Mice , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , Transcription, Genetic/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/metabolism , Ubiquitination
14.
Mol Cell ; 36(6): 954-69, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-20064462

ABSTRACT

In response to DNA double-strand breaks (DSBs), cells sense the DNA lesions and then activate the protein kinase ATM. Subsequent DSB resection produces RPA-coated ssDNA that is essential for activation of the DNA damage checkpoint and DNA repair by homologous recombination (HR). However, the biochemical mechanism underlying the transition from DSB sensing to resection remains unclear. Using Xenopus egg extracts and human cells, we show that the tumor suppressor protein CtIP plays a critical role in this transition. We find that CtIP translocates to DSBs, a process dependent on the DSB sensor complex Mre11-Rad50-NBS1, the kinase activity of ATM, and a direct DNA-binding motif in CtIP, and then promotes DSB resection. Thus, CtIP facilitates the transition from DSB sensing to processing: it does so by binding to the DNA at DSBs after DSB sensing and ATM activation and then promoting DNA resection, leading to checkpoint activation and HR.


Subject(s)
Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases , Enzyme Activation , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic , Sequence Alignment , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenopus laevis
15.
Proc Natl Acad Sci U S A ; 111(13): E1240-8, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24639535

ABSTRACT

Breast cancer susceptibility gene 1 (BRCA1) is a breast and ovarian cancer tumor suppressor whose loss leads to DNA damage and defective centrosome functions. Despite its tumor suppression functions, BRCA1 is most highly expressed in the embryonic neuroepithelium when the neural progenitors are highly proliferative. To determine its functional significance, we deleted BRCA1 in the developing brain using a neural progenitor-specific driver. The phenotype is characterized by severe agenesis of multiple laminated cerebral structures affecting most notably the neocortex, hippocampus, cerebellum, and olfactory bulbs. Major phenotypes are caused by excess apoptosis, as these could be significantly suppressed by the concomitant deletion of p53. Certain phenotypes attributable to centrosomal and cell polarity functions could not be rescued by p53 deletion. A double KO with the DNA damage sensor kinase ATM was able to rescue BRCA1 loss to a greater extent than p53. Our results suggest distinct apoptotic and centrosomal functions of BRCA1 in neural progenitors, with important implications to understand the sensitivity of the embryonic brain to DNA damage, as well as the developmental regulation of brain size.


Subject(s)
BRCA1 Protein/metabolism , Brain/embryology , Brain/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Brain/cytology , Cell Polarity , Cell Proliferation , Cell Survival , Cognition/physiology , Gene Deletion , Learning , Mice , Mice, Knockout , Motor Activity/physiology , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phenotype , Signal Transduction , Tumor Suppressor Protein p53/metabolism
16.
Proc Natl Acad Sci U S A ; 111(17): E1723-30, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24706852

ABSTRACT

Despite therapeutic advancement, pulmonary disease still remains a major cause of morbidity and mortality around the world. Opportunities to study human lung disease either in vivo or in vitro are currently limited. Using induced pluripotent stem cells (iPSCs), we generated mature multiciliated cells in a functional airway epithelium. Robust multiciliogenesis occurred when notch signaling was inhibited and was confirmed by (i) the assembly of multiple pericentrin-stained centrioles at the apical surface, (ii) expression of transcription factor forkhead box protein J1, and (iii) presence of multiple acetylated tubulin-labeled cilia projections in individual cells. Clara, goblet, and basal cells were all present, confirming the generation of a complete polarized epithelial-cell layer. Additionally, cAMP-activated and cystic fibrosis transmembrane regulator inhibitor 172-sensitive cystic fibrosis transmembrane regulator currents were recorded in isolated epithelial cells. Our report demonstrating the generation of mature multiciliated cells in respiratory epithelium from iPSCs is a significant advance toward modeling a number of human respiratory diseases in vitro.


Subject(s)
Cilia/metabolism , Epithelial Cells/cytology , Epithelium/metabolism , Induced Pluripotent Stem Cells/cytology , Lung/cytology , Cell Differentiation , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoderm/cytology , Epithelial Cells/metabolism , Humans , Receptors, Notch/metabolism , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 111(32): E3343-52, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071179

ABSTRACT

Glial cells are an integral part of functional communication in the brain. Here we show that astrocytes contribute to the fast dynamics of neural circuits that underlie normal cognitive behaviors. In particular, we found that the selective expression of tetanus neurotoxin (TeNT) in astrocytes significantly reduced the duration of carbachol-induced gamma oscillations in hippocampal slices. These data prompted us to develop a novel transgenic mouse model, specifically with inducible tetanus toxin expression in astrocytes. In this in vivo model, we found evidence of a marked decrease in electroencephalographic (EEG) power in the gamma frequency range in awake-behaving mice, whereas neuronal synaptic activity remained intact. The reduction in cortical gamma oscillations was accompanied by impaired behavioral performance in the novel object recognition test, whereas other forms of memory, including working memory and fear conditioning, remained unchanged. These results support a key role for gamma oscillations in recognition memory. Both EEG alterations and behavioral deficits in novel object recognition were reversed by suppression of tetanus toxin expression. These data reveal an unexpected role for astrocytes as essential contributors to information processing and cognitive behavior.


Subject(s)
Astrocytes/physiology , Recognition, Psychology/physiology , Animals , Astrocytes/drug effects , Brain Waves/drug effects , Brain Waves/physiology , Calcium Signaling , Carbachol/pharmacology , Electroencephalography , Gene Expression , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nerve Net/cytology , Nerve Net/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Tissue Culture Techniques , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
18.
PLoS Biol ; 11(6): e1001581, 2013.
Article in English | MEDLINE | ID: mdl-23776406

ABSTRACT

Activation of the IκB kinase (IKK) is central to NF-κB signaling. However, the precise activation mechanism by which catalytic IKK subunits gain the ability to induce NF-κB transcriptional activity is not well understood. Here we report a 4 Å x-ray crystal structure of human IKK2 (hIKK2) in its catalytically active conformation. The hIKK2 domain architecture closely resembles that of Xenopus IKK2 (xIKK2). However, whereas inactivated xIKK2 displays a closed dimeric structure, hIKK2 dimers adopt open conformations that permit higher order oligomerization within the crystal. Reversible oligomerization of hIKK2 dimers is observed in solution. Mutagenesis confirms that two of the surfaces that mediate oligomerization within the crystal are also critical for the process of hIKK2 activation in cells. We propose that IKK2 dimers transiently associate with one another through these interaction surfaces to promote trans auto-phosphorylation as part of their mechanism of activation. This structure-based model supports recently published structural data that implicate strand exchange as part of a mechanism for IKK2 activation via trans auto-phosphorylation. Moreover, oligomerization through the interfaces identified in this study and subsequent trans auto-phosphorylation account for the rapid amplification of IKK2 phosphorylation observed even in the absence of any upstream kinase.


Subject(s)
I-kappa B Kinase/chemistry , I-kappa B Kinase/metabolism , Chromatography, Gel , Crystallography, X-Ray , Enzyme Activation , HEK293 Cells , Humans , Models, Molecular , Phosphorylation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Solutions , Structure-Activity Relationship , Transfection
19.
EMBO Rep ; 15(3): 244-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24531722

ABSTRACT

Regenerative medicine aims to replace the lost or damaged cells in the human body through a new source of healthy transplanted cells or by endogenous repair. Although human embryonic stem cells were first thought to be the ideal source for cell therapy and tissue repair in humans, the discovery by Yamanaka and colleagues revolutionized the field. Almost any differentiated cell can be sent back in time to a pluripotency state by expressing the appropriate transcription factors. The process of somatic reprogramming using Yamanaka factors, many of which are oncogenes, offers a glimpse into how cancer stem cells may originate. In this review we discuss the similarities between tumor dedifferentiation and somatic cell reprogramming and how this may pose a risk to the application of this new technology in regenerative medicine.


Subject(s)
Cell Dedifferentiation , Cellular Reprogramming , Neoplastic Stem Cells/cytology , Animals , Carcinogens/metabolism , Cell Lineage , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology
20.
Immunol Rev ; 246(1): 239-53, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22435559

ABSTRACT

The inhibitor of nuclear factor-κB (IκB) kinase (IKK) complex is the master regulator of the NF-κB signaling pathway. The activation of the IKK complex is a tightly regulated, highly stimulus-specific, and target-specific event that is essential for the plethora of functions attributed to NF-κB. More recently, NF-κB-independent roles of IKK members have brought increased complexity to its biological function. This review highlights some of the major advances in the studies of the process of IKK activation and the biological roles of IKK family members, with a focus on NF-κB-independent functions. Understanding these complex processes is essential for targeting IKK for therapeutics.


Subject(s)
I-kappa B Kinase/metabolism , Animals , Enzyme Activation , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL