Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nature ; 589(7840): 110-115, 2021 01.
Article in English | MEDLINE | ID: mdl-33239785

ABSTRACT

In mammals, telomere protection is mediated by the essential protein TRF2, which binds chromosome ends and ensures genome integrity1,2. TRF2 depletion results in end-to-end chromosome fusions in all cell types that have been tested so far. Here we find that TRF2 is dispensable for the proliferation and survival of mouse embryonic stem (ES) cells. Trf2-/- (also known as Terf2) ES cells do not exhibit telomere fusions and can be expanded indefinitely. In response to the deletion of TRF2, ES cells exhibit a muted DNA damage response that is characterized by the recruitment of γH2AX-but not 53BP1-to telomeres. To define the mechanisms that control this unique DNA damage response in ES cells, we performed a CRISPR-Cas9-knockout screen. We found a strong dependency of TRF2-null ES cells on the telomere-associated protein POT1B and on the chromatin remodelling factor BRD2. Co-depletion of POT1B or BRD2 with TRF2 restores a canonical DNA damage response at telomeres, resulting in frequent telomere fusions. We found that TRF2 depletion in ES cells activates a totipotent-like two-cell-stage transcriptional program that includes high levels of ZSCAN4. We show that the upregulation of ZSCAN4 contributes to telomere protection in the absence of TRF2. Together, our results uncover a unique response to telomere deprotection during early development.


Subject(s)
Pluripotent Stem Cells/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/deficiency , Telomeric Repeat Binding Protein 2/metabolism , Animals , Cell Proliferation , Cell Survival , DNA Damage , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Telomeric Repeat Binding Protein 2/genetics , Totipotent Stem Cells/cytology , Totipotent Stem Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Cell ; 144(5): 782-95, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376238

ABSTRACT

During development and regeneration, proliferation of tissue-specific stem cells is tightly controlled to produce organs of a predetermined size. The molecular determinants of this process remain poorly understood. Here, we investigate the function of Yap1, the transcriptional effector of the Hippo signaling pathway, in skin biology. Using gain- and loss-of-function studies, we show that Yap1 is a critical modulator of epidermal stem cell proliferation and tissue expansion. Yap1 mediates this effect through interaction with TEAD transcription factors. Additionally, our studies reveal that α-catenin, a molecule previously implicated in tumor suppression and cell density sensing in the skin, is an upstream negative regulator of Yap1. α-catenin controls Yap1 activity and phosphorylation by modulating its interaction with 14-3-3 and the PP2A phosphatase. Together, these data identify Yap1 as a determinant of the proliferative capacity of epidermal stem cells and as an important effector of a "crowd control" molecular circuitry in mammalian skin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Epidermal Cells , Phosphoproteins/metabolism , alpha Catenin/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Cycle Proteins , Cell Line , Epidermis/metabolism , Mice , YAP-Signaling Proteins
3.
Int J Mol Sci ; 21(16)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796510

ABSTRACT

Autoimmune diabetes is a complex multifactorial disease with genetic and environmental factors playing pivotal roles. While many genes associated with the risk of diabetes have been identified to date, the mechanisms by which external triggers contribute to the genetic predisposition remain unclear. Here, we derived embryonic stem (ES) cell lines from diabetes-prone non-obese diabetic (NOD) and healthy C57BL/6 (B6) mice. While overall pluripotency markers were indistinguishable between newly derived NOD and B6 ES cells, we discovered several differentially expressed genes that normally are not expressed in ES cells. Several genes that reside in previously identified insulin-dependent diabetics (Idd) genomic regions were up-regulated in NOD ES cells. Gene set enrichment analysis showed that different groups of genes associated with immune functions are differentially expressed in NOD. Transcriptomic analysis of NOD blastocysts validated several differentially overexpressed Idd genes compared to B6. Genome-wide mapping of active histone modifications using ChIP-Seq supports active expression as the promoters and enhancers of activated genes are also marked by active histone modifications. We have also found that NOD ES cells secrete more inflammatory cytokines. Our data suggest that the known genetic predisposition of NOD to autoimmune diabetes leads to epigenetic instability of several Idd regions.


Subject(s)
Autoimmunity/genetics , Blastocyst/metabolism , Immune System/metabolism , Mouse Embryonic Stem Cells/metabolism , Transcription, Genetic , Animals , Chemokines/metabolism , Chromatin/metabolism , Diabetes Mellitus, Experimental/genetics , Epigenesis, Genetic , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Proteome/metabolism , Proteomics , Transcriptome/genetics
4.
Nature ; 493(7434): 679-83, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23263183

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) pathway regulates organismal growth in response to many environmental cues, including nutrients and growth factors. Cell-based studies showed that mTORC1 senses amino acids through the RagA-D family of GTPases (also known as RRAGA, B, C and D), but their importance in mammalian physiology is unknown. Here we generate knock-in mice that express a constitutively active form of RagA (RagA(GTP)) from its endogenous promoter. RagA(GTP/GTP) mice develop normally, but fail to survive postnatal day 1. When delivered by Caesarean section, fasted RagA(GTP/GTP) neonates die almost twice as rapidly as wild-type littermates. Within an hour of birth, wild-type neonates strongly inhibit mTORC1, which coincides with profound hypoglycaemia and a decrease in plasma amino-acid concentrations. In contrast, mTORC1 inhibition does not occur in RagA(GTP/GTP) neonates, despite identical reductions in blood nutrient amounts. With prolonged fasting, wild-type neonates recover their plasma glucose concentrations, but RagA(GTP/GTP) mice remain hypoglycaemic until death, despite using glycogen at a faster rate. The glucose homeostasis defect correlates with the inability of fasted RagA(GTP/GTP) neonates to trigger autophagy and produce amino acids for de novo glucose production. Because profound hypoglycaemia does not inhibit mTORC1 in RagA(GTP/GTP) neonates, we considered the possibility that the Rag pathway signals glucose as well as amino-acid sufficiency to mTORC1. Indeed, mTORC1 is resistant to glucose deprivation in RagA(GTP/GTP) fibroblasts, and glucose, like amino acids, controls its recruitment to the lysosomal surface, the site of mTORC1 activation. Thus, the Rag GTPases signal glucose and amino-acid concentrations to mTORC1, and have an unexpectedly key role in neonates in autophagy induction and thus nutrient homeostasis and viability.


Subject(s)
Animals, Newborn/physiology , Autophagy/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Enzymologic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Amino Acids/metabolism , Animals , Animals, Newborn/metabolism , Blood Glucose/metabolism , GTP Phosphohydrolases/genetics , Gene Knock-In Techniques , Hypoglycemia/genetics , Kaplan-Meier Estimate , Mechanistic Target of Rapamycin Complex 1 , Mice , Time Factors
5.
Proc Natl Acad Sci U S A ; 113(16): E2316-25, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044095

ABSTRACT

To study the development and function of "natural-arising" T regulatory (nTreg) cells, we developed a novel nTreg model on pure nonobese diabetic background using epigenetic reprogramming via somatic cell nuclear transfer. On RAG1-deficient background, we found that monoclonal FoxP3(+)CD4(+)Treg cells developed in the thymus in the absence of other T cells. Adoptive transfer experiments revealed that the thymic niche is not a limiting factor in nTreg development. In addition, we showed that the T-cell receptor (TCR) ß-chain of our nTreg model was not only sufficient to bias T-cell development toward the CD4 lineage, but we also demonstrated that this TCR ß-chain was able to provide stronger TCR signals. This TCR-ß-driven mechanism would thus unify former per se contradicting hypotheses of TCR-dependent and -independent nTreg development. Strikingly, peripheral FoxP3(-)CD4(+)T cells expressing the same TCR as this somatic cell nuclear transfer nTreg model had a reduced capability to differentiate into Th1 cells but were poised to differentiate better into induced nTreg cells, both in vitro and in vivo, representing a novel peripheral precursor subset of nTreg cells to which we refer to as pre-nTreg cells.


Subject(s)
Cell Differentiation/immunology , Models, Immunological , Nuclear Transfer Techniques , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology
6.
Blood ; 121(7): 1145-56, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23243279

ABSTRACT

Antigen presenting cells (APCs) that express a catalytically inactive version of the deubiquitylase YOD1 (YOD1-C160S) present exogenous antigens more efficiently to CD8(+) T cells, both in vitro and in vivo. Compared with controls, immunization of YOD1-C160S mice led to greater expansion of specific CD8(+) T cells and showed improved control of infection with a recombinant -herpes virus, MHV-68, engineered to express SIINFEKL peptide, the ligand for the ovalbumin-specific TCR transgenic OT-I cells. Enhanced expansion of specific CD8(+) T cells was likewise observed on infection of YOD1-C160S mice with a recombinant influenza A virus expressing SIINFEKL. YOD1-C160S APCs retained antigen longer than did control APCs. Enhanced crosspresentation by YOD1-C160S APCs was transporter associated with antigen processing (TAP1)-independent but sensitive to inclusion of inhibitors of acidification and of the proteasome. The activity of deubiquitylating enzymes may thus help control antigenspecific CD8(+) T-cell responses during immunization.


Subject(s)
Cross-Priming , Mutation, Missense , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Brefeldin A/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/drug effects , Cross-Priming/genetics , Female , Hydrogen-Ion Concentration , Immunization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Rhadinovirus/immunology , Rhadinovirus/pathogenicity , Ubiquitin Thiolesterase/metabolism
7.
J Immunol ; 190(2): 695-702, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23241879

ABSTRACT

Recognition of nucleic acids by TLR9 requires its trafficking from the endoplasmic reticulum to endolysosomal compartments and its subsequent proteolytic processing. Both processes depend on interactions of TLR9 with the polytopic endoplasmic reticulum-resident protein UNC93B1. To examine the intracellular behavior of TLR9 in primary APCs, we generated transgenic mice expressing a TLR9-GFP fusion. The TLR9-GFP transgene is functional and is proteolytically processed in resting bone marrow-derived macrophages (BMDMs), dendritic cells, and B cells. Inhibition of cleavage impairs TLR9-dependent responses in all primary APCs analyzed. The kinetics of TLR9-GFP processing in BMDMs and B cells differs: in B cells, proteolysis occurs at a faster rate, consistent with an almost exclusive localization to endolysosomes at the resting state. In contrast to the joint requirement for cathepsins L and S for TLR9 cleavage in macrophages, TLR9-GFP cleavage depends on cathepsin L activity in B cells. As expected, in BMDMs and B cells from UNC93B1 (3d) mutant mice, cleavage of TLR9-GFP is essentially blocked, and the expression level of UNC93B1 appears tightly correlated with TLR9-GFP cleavage. We conclude that proteolysis is a universal requirement for TLR9 activation in the primary cell types tested, however the cathepsin requirement, rate of cleavage, and intracellular behavior of TLR9 varies. The observed differences in trafficking indicate the possibility of distinct modes of endosomal content sampling to facilitate initiation of TLR-driven responses in APCs.


Subject(s)
Antigen-Presenting Cells/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Animals , B-Lymphocytes/metabolism , Bone Marrow Cells/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lysosomes/metabolism , Macrophages/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Transgenic , Protein Stability , Protein Transport , Proteolysis , Signal Transduction , Transgenes
8.
PLoS Biol ; 8(3): e1000605, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21468303

ABSTRACT

Ubiquitin-dependent processes control much of cellular physiology. We show that expression of a highly active, Epstein-Barr virus-derived deubiquitylating enzyme (EBV-DUB) blocks proteasomal degradation of cytosolic and ER-derived proteins by preemptive removal of ubiquitin from proteasome substrates, a treatment less toxic than the use of proteasome inhibitors. Recognition of misfolded proteins in the ER lumen, their dislocation to the cytosol, and degradation are usually tightly coupled but can be uncoupled by the EBV-DUB: a misfolded glycoprotein that originates in the ER accumulates in association with cytosolic chaperones as a deglycosylated intermediate. Our data underscore the necessity of a DUB activity for completion of the dislocation reaction and provide a new means of inhibition of proteasomal proteolysis with reduced cytotoxicity.


Subject(s)
Herpesvirus 4, Human/enzymology , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism , Viral Proteins/metabolism , Biocatalysis , Cell Line , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Humans , Molecular Chaperones/metabolism , Protein Folding , Protein Processing, Post-Translational , Protein Transport , Substrate Specificity
9.
Nature ; 451(7182): 1125-9, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18278031

ABSTRACT

MicroRNAs are abundant in animal genomes and have been predicted to have important roles in a broad range of gene expression programmes. Despite this prominence, there is a dearth of functional knowledge regarding individual mammalian microRNAs. Using a loss-of-function allele in mice, we report here that the myeloid-specific microRNA-223 (miR-223) negatively regulates progenitor proliferation and granulocyte differentiation and activation. miR-223 (also called Mirn223) mutant mice have an expanded granulocytic compartment resulting from a cell-autonomous increase in the number of granulocyte progenitors. We show that Mef2c, a transcription factor that promotes myeloid progenitor proliferation, is a target of miR-223, and that genetic ablation of Mef2c suppresses progenitor expansion and corrects the neutrophilic phenotype in miR-223 null mice. In addition, granulocytes lacking miR-223 are hypermature, hypersensitive to activating stimuli and display increased fungicidal activity. As a consequence of this neutrophil hyperactivity, miR-223 mutant mice spontaneously develop inflammatory lung pathology and exhibit exaggerated tissue destruction after endotoxin challenge. Our data support a model in which miR-223 acts as a fine-tuner of granulocyte production and the inflammatory response.


Subject(s)
Cell Proliferation , Granulocytes/cytology , Granulocytes/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Stem Cells/cytology , Alleles , Animals , Cell Differentiation , Gene Deletion , Granulocytes/immunology , Granulocytes/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lung/pathology , MEF2 Transcription Factors , Mice , Mice, Knockout , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Neutrophils/physiology , Phenotype
10.
PLoS Genet ; 7(5): e1002054, 2011 May.
Article in English | MEDLINE | ID: mdl-21573140

ABSTRACT

MicroRNAs (miRNAs) post-transcriptionally regulate the expression of thousands of distinct mRNAs. While some regulatory interactions help to maintain basal cellular functions, others are likely relevant in more specific settings, such as response to stress. Here we describe such a role for the mir-290-295 cluster, the dominant miRNA cluster in mouse embryonic stem cells (mESCs). Examination of a target list generated from bioinformatic prediction, as well as expression data following miRNA loss, revealed strong enrichment for apoptotic regulators, two of which we validated directly: Caspase 2, the most highly conserved mammalian caspase, and Ei24, a p53 transcriptional target. Consistent with these predictions, mESCs lacking miRNAs were more likely to initiate apoptosis following genotoxic exposure to gamma irradiation or doxorubicin. Knockdown of either candidate partially rescued this pro-apoptotic phenotype, as did transfection of members of the mir-290-295 cluster. These findings were recapitulated in a specific mir-290-295 deletion line, confirming that they reflect miRNA functions at physiological levels. In contrast to the basal regulatory roles previously identified, the pro-survival phenotype shown here may be most relevant to stressful gestations, where pro-oxidant metabolic states induce DNA damage. Similarly, this cluster may mediate chemotherapeutic resistance in a neoplastic context, making it a useful clinical target.


Subject(s)
Embryonic Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Caspase 2/genetics , Caspase 2/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/drug effects , DNA Damage/radiation effects , Doxorubicin/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/radiation effects , Gamma Rays , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 108(34): 14163-8, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21844366

ABSTRACT

Mir-290 through mir-295 (mir-290-295) is a mammalian-specific microRNA (miRNA) cluster that, in mice, is expressed specifically in early embryos and embryonic germ cells. Here, we show that mir-290-295 plays important roles in embryonic development as indicated by the partially penetrant lethality of mutant embryos. In addition, we show that in surviving mir-290-295-deficient embryos, female but not male fertility is compromised. This impairment in fertility arises from a defect in migrating primordial germ cells and occurs equally in male and female mutant animals. Male mir-290-295(-/-) mice, due to the extended proliferative lifespan of their germ cells, are able to recover from this initial germ cell loss and are fertile. Female mir-290-295(-/-) mice are unable to recover and are sterile, due to premature ovarian failure.


Subject(s)
Embryo Loss/genetics , Embryo Loss/pathology , Germ Cells/metabolism , Germ Cells/pathology , MicroRNAs/metabolism , Penetrance , Aging/pathology , Animals , Animals, Newborn , Apoptosis , Cell Count , Cell Cycle , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fertility/genetics , Gene Expression Regulation, Developmental , Gonads/growth & development , Gonads/pathology , Infertility, Female/genetics , Infertility, Female/pathology , Male , Mice , Mice, Mutant Strains , MicroRNAs/genetics
13.
Nat Methods ; 6(2): 147-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19122668

ABSTRACT

Cell fusion has been used for many different purposes, including generation of hybridomas and reprogramming of somatic cells. The fusion step is the key event in initiation of these procedures. Standard fusion techniques, however, provide poor and random cell contact, leading to low yields. We present here a microfluidic device to trap and properly pair thousands of cells. Using this device, we paired different cell types, including fibroblasts, mouse embryonic stem cells and myeloma cells, achieving pairing efficiencies up to 70%. The device is compatible with both chemical and electrical fusion protocols. We observed that electrical fusion was more efficient than chemical fusion, with membrane reorganization efficiencies of up to 89%. We achieved greater than 50% properly paired and fused cells over the entire device, fivefold greater than with a commercial electrofusion chamber and observed reprogramming in hybrids between mouse embryonic stem cells and mouse embryonic fibroblasts.


Subject(s)
Cell Fusion/methods , Microfluidic Analytical Techniques/instrumentation , Animals , Cell Line, Tumor , Electroporation/methods , Embryonic Stem Cells , Image Processing, Computer-Assisted/methods , Mice , Microfluidic Analytical Techniques/methods , NIH 3T3 Cells , Polyethylene Glycols/pharmacology
14.
Stem Cells ; 29(6): 992-1000, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21563275

ABSTRACT

Pluripotent cells can be derived from different types of somatic cells by nuclear reprogramming through the ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc. However, it is unclear whether postmitotic neurons are susceptible to direct reprogramming. Here, we show that postnatal cortical neurons, the vast majority of which are postmitotic, are amenable to epigenetic reprogramming. However, ectopic expression of the four canonical reprogramming factors is not sufficient to reprogram postnatal neurons. Efficient reprogramming was only achieved after forced cell proliferation by p53 suppression. Additionally, overexpression of repressor element-1 silencing transcription, a suppressor of neuronal gene activity, increased reprogramming efficiencies in combination with the reprogramming factors. Our findings indicate that terminally differentiated postnatal neurons are able to acquire the pluripotent state by direct epigenetic reprogramming, and this process is made more efficient through the suppression of lineage specific gene expression.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Repressor Proteins/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Blastocyst/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Coculture Techniques , Embryo Transfer , Fibroblasts/cytology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Lewis X Antigen/metabolism , Mice , Nanog Homeobox Protein , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , Teratoma/pathology , Transplantation Chimera
16.
Front Immunol ; 11: 1073, 2020.
Article in English | MEDLINE | ID: mdl-32625203

ABSTRACT

While B cells play a significant role in the onset of type-1 diabetes (T1D), little is know about their role in those early stages. Thus, to gain new insights into the role of B cells in T1D, we converted a physiological early pancreas-infiltrating B cell into a novel BCR mouse model using Somatic Cell Nuclear Transfer (SCNT). Strikingly, SCNT-derived B1411 model displayed neither developmental block nor anergy. Instead, B1411 underwent spontaneous germinal center reactions. Without T cell help, B1411-Rag1-/- was capable of forming peri-/intra-pancreatic lymph nodes, and undergoing class-switching. RNA-Seq analysis identified 93 differentially expressed genes in B1411 compared to WT B cells, including Irf7, Usp18, and Mda5 that had been linked to a potential viral etiology of T1D. We also found various members of the oligoadenylate synthase (OAS) family to be enriched in B1411, such as Oas1, which had recently also been linked to T1D. Strikingly, when challenged with glucose B1411-Rag1-/- mice displayed impaired glucose tolerance.


Subject(s)
Autoimmunity , B-Lymphocytes/immunology , Prediabetic State/etiology , Prediabetic State/immunology , Animals , Basidiomycota/genetics , Basidiomycota/metabolism , Calcium Signaling/immunology , Chromatin Assembly and Disassembly , Clone Cells/immunology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Female , Gene Expression Profiling , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Knockout , Models, Immunological , Nuclear Transfer Techniques , Prediabetic State/genetics , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology
17.
Stem Cells ; 26(6): 1628-35, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18369099

ABSTRACT

Successful hematopoietic stem cell (HSC) transplantation is often limited by the numbers of HSCs, and robust methods to expand HSCs ex vivo are needed. We previously showed that angiopoietin-like proteins (Angptls), a group of growth factors isolated from a fetal liver HSC-supportive cell population, improved ex vivo expansion of HSCs. Here, we demonstrate that insulin-like growth factor-binding protein 2 (IGFBP2), secreted by a tumorigenic cell line, also enhanced ex vivo expansion of mouse HSCs. On the basis of these findings, we established a completely defined, serum-free culture system for mouse HSCs, containing SCF, thrombopoietin, fibroblast growth factor 1, Angptl3, and IGFBP2. As measured by competitive repopulation analyses, there was a 48-fold increase in numbers of long-term repopulating mouse HSCs after 21 days of culture. This is the first demonstration that IGFBP2 stimulates expansion or proliferation of murine stem cells. Our finding also suggests that certain cancer cells synthesize proteins that can stimulate HSC expansion. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Hematopoietic Stem Cells/cytology , Insulin-Like Growth Factor Binding Protein 1/pharmacology , Tissue Expansion/methods , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Line , Cell Line, Tumor , Culture Media, Conditioned , Hematopoietic Stem Cells/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 1/metabolism , Kidney , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transfection
18.
Dev Cell ; 29(3): 321-9, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24768164

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) integrates cues from growth factors and nutrients to control metabolism. In contrast to the growth factor input, genetic disruption of nutrient-dependent activation of mTORC1 in mammals remains unexplored. We engineered mice lacking RagA and RagB genes, which encode the GTPases responsible for mTORC1 activation by nutrients. RagB has limited expression, and its loss shows no effects on mammalian physiology. RagA deficiency leads to E10.5 embryonic death, loss of mTORC1 activity, and severe growth defects. Primary cells derived from these mice exhibit no regulation of mTORC1 by nutrients and maintain high sensitivity to growth factors. Deletion of RagA in adult mice is lethal. Upon RagA loss, a myeloid population expands in peripheral tissues. RagA-specific deletion in liver increases cellular responses to growth factors. These results show the essentiality of nutrient sensing for mTORC1 activity in mice and its suppression of PI3K/Akt signaling.


Subject(s)
Embryo, Mammalian/embryology , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Hepatocytes/metabolism , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
19.
Cell Rep ; 1(5): 461-71, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22832272

ABSTRACT

To study the CD8(+) T cell response against a mouse γ-herpes virus, we generated K(b)-MHV-68-ORF8(604-612)RAG(-/-) CD8(+) T cell receptor transnuclear (TN) mice as a source of virus-specific CD8(+) T cells. K(b)-ORF8-Tet(+) CD8(+) T cells, expanded in the course of a resolving MHV-68 infection, served as a source of nucleus donors. Various in vivo and ex vivo assay criteria demonstrated the fine specificity and functionality of TN cells. TN cells proliferated extensively in response to viral infection, helped control viral burden, and exhibited a phenotype similar to that of endogenous K(b)-ORF8-Tet(+) cells. When compared to OT-1 cells, TN cells displayed distinct properties in response to lymphopenia and cognate antigen stimulation, which may be attributable to the affinity of the TCR expressed by the TN cells. The availability of MHV-68-specific CD8(+) TCR TN mice provides a new tool for investigating aspects of host-pathogen interactions unique to γ-herpes viruses.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Epitopes/metabolism , Glycoproteins/metabolism , H-2 Antigens/metabolism , Herpesviridae Infections/physiopathology , Receptors, Antigen, T-Cell/metabolism , Rhadinovirus/metabolism , Viral Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Herpesviridae Infections/metabolism , Herpesviridae Infections/prevention & control , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Phenotype , Tumor Virus Infections/metabolism , Tumor Virus Infections/physiopathology , Tumor Virus Infections/prevention & control , Viral Load/physiology
20.
PLoS One ; 6(4): e18817, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21533087

ABSTRACT

Controlled localization of class II MHC molecules is essential for proper class II MHC-restricted antigen presentation and the subsequent initiation of an adaptive immune response. Ubiquitination of class II MHC molecules on cytosolic lysine (K225) of the ß-chain has been shown to affect localization of the complex. We generated mice in which the endogenous ß-chain locus is replaced with a GFP tagged mutant version that lacks the cytosolic lysine residue (I-A-ß-K225R-EGFP). These mice have elevated levels of class II MHC as compared to I-A-ß-EGFP mice, and immature bone marrow-derived dendritic cells show redistribution of class II MHC to the cell surface. Nonetheless, in these same cells efficiency of antigen presentation is unaffected in I-A-ß-K225R-EGFP mice, as assayed for presentation of ovalbumin to appropriately specific T cells. The I-A-ß-K225R-EGFP animals have normal CD4 T cell populations and are capable of generating antigen-specific antibody in response to model antigens and viral infection. We therefore conclude that in our experimental system modulation of trafficking by ubiquitination of residue K225 of the ß-chain is not essential for the function of class II MHC products in antigen presentation or antibody production.


Subject(s)
Antibody Formation , Antigens/biosynthesis , Histocompatibility Antigens Class II/metabolism , Ubiquitin/physiology , Animals , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL