Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Nat Commun ; 12(1): 4217, 2021 07 09.
Article En | MEDLINE | ID: mdl-34244513

The functional consequences of genetic variants within 5' untranslated regions (UTRs) on a genome-wide scale are poorly understood in disease. Here we develop a high-throughput multi-layer functional genomics method called PLUMAGE (Pooled full-length UTR Multiplex Assay on Gene Expression) to quantify the molecular consequences of somatic 5' UTR mutations in human prostate cancer. We show that 5' UTR mutations can control transcript levels and mRNA translation rates through the creation of DNA binding elements or RNA-based cis-regulatory motifs. We discover that point mutations can simultaneously impact transcript and translation levels of the same gene. We provide evidence that functional 5' UTR mutations in the MAP kinase signaling pathway can upregulate pathway-specific gene expression and are associated with clinical outcomes. Our study reveals the diverse mechanisms by which the mutational landscape of 5' UTRs can co-opt gene expression and demonstrates that single nucleotide alterations within 5' UTRs are functional in cancer.


5' Untranslated Regions/genetics , DNA Mutational Analysis/methods , Gene Expression Regulation, Neoplastic , Genomics/methods , Prostatic Neoplasms/genetics , Cell Line, Tumor , HEK293 Cells , High-Throughput Screening Assays , Humans , Male , Point Mutation , Prostate/pathology , Prostatic Neoplasms/pathology , Protein Biosynthesis/genetics , RNA-Seq
2.
PLoS Genet ; 16(12): e1009186, 2020 12.
Article En | MEDLINE | ID: mdl-33306674

Cells are exposed to frequent mechanical and/or chemical stressors that can compromise the integrity of the plasma membrane and underlying cortical cytoskeleton. The molecular mechanisms driving the immediate repair response launched to restore the cell cortex and circumvent cell death are largely unknown. Using microarrays and drug-inhibition studies to assess gene expression, we find that initiation of cell wound repair in the Drosophila model is dependent on translation, whereas transcription is required for subsequent steps. We identified 253 genes whose expression is up-regulated (80) or down-regulated (173) in response to laser wounding. A subset of these genes were validated using RNAi knockdowns and exhibit aberrant actomyosin ring assembly and/or actin remodeling defects. Strikingly, we find that the canonical insulin signaling pathway controls actin dynamics through the actin regulators Girdin and Chickadee (profilin), and its disruption leads to abnormal wound repair. Our results provide new insight for understanding how cell wound repair proceeds in healthy individuals and those with diseases involving wound healing deficiencies.


Actins/metabolism , Autocrine Communication , Insulin/metabolism , Signal Transduction , Wound Healing , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Profilins/genetics , Profilins/metabolism , Transcriptome
3.
Nat Commun ; 10(1): 4596, 2019 10 10.
Article En | MEDLINE | ID: mdl-31601799

Many of the regulatory features governing erythrocyte specification, maturation, and associated disorders remain enigmatic. To identify new regulators of erythropoiesis, we utilize a functional genomic screen for genes affecting expression of the erythroid marker CD235a/GYPA. Among validating hits are genes coding for the N6-methyladenosine (m6A) mRNA methyltransferase (MTase) complex, including, METTL14, METTL3, and WTAP. We demonstrate that m6A MTase activity promotes erythroid gene expression programs through selective translation of ~300 m6A marked mRNAs, including those coding for SETD histone methyltransferases, ribosomal components, and polyA RNA binding proteins. Remarkably, loss of m6A marks results in dramatic loss of H3K4me3 marks across key erythroid-specific KLF1 transcriptional targets (e.g., Heme biosynthesis genes). Further, each m6A MTase subunit and a subset of their mRNAs targets are required for human erythroid specification in primary bone-marrow derived progenitors. Thus, m6A mRNA marks promote the translation of a network of genes required for human erythropoiesis.


Adenosine/analogs & derivatives , Erythropoiesis/genetics , Protein Biosynthesis , Adenosine/genetics , Antigens, CD34/genetics , Antigens, CD34/metabolism , Bone Marrow Cells/physiology , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia, Erythroblastic, Acute/genetics , Methyltransferases/genetics , Promoter Regions, Genetic , RNA Splicing Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulon
4.
Nat Med ; 25(10): 1566-1575, 2019 10.
Article En | MEDLINE | ID: mdl-31591594

The ability to expand hematopoietic stem and progenitor cells (HSPCs) ex vivo is critical to fully realize the potential of HSPC-based therapies. In particular, the application of clinically effective therapies, such as cord blood transplantation, has been impeded because of limited HSPC availability. Here, using 3D culture of human HSPCs in a degradable zwitterionic hydrogel, we achieved substantial expansion of phenotypically primitive CD34+ cord blood and bone-marrow-derived HSPCs. This culture system led to a 73-fold increase in long-term hematopoietic stem cell (LT-HSC) frequency, as demonstrated by limiting dilution assays, and the expanded HSPCs were capable of hematopoietic reconstitution for at least 24 weeks in immunocompromised mice. Both the zwitterionic characteristics of the hydrogel and the 3D format were important for HSPC self-renewal. Mechanistically, the impact of 3D zwitterionic hydrogel culture on mitigating HSPC differentiation and promoting self-renewal might result from an inhibition of excessive reactive oxygen species (ROS) production via suppression of O2-related metabolism. HSPC expansion using zwitterionic hydrogels has the potential to facilitate the clinical application of hematopoietic-stem-cell therapies.


Cell Differentiation/drug effects , Cell- and Tissue-Based Therapy , Hematopoietic Stem Cells/cytology , Hydrogels/pharmacology , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Culture Techniques , Cell Proliferation/drug effects , Coculture Techniques , Fetal Blood/cytology , Fetal Blood/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Mice , Reactive Oxygen Species/metabolism
5.
Genome Med ; 11(1): 14, 2019 03 12.
Article En | MEDLINE | ID: mdl-30867038

It was highlighted that in the original article [1] the Availability of data and materials section was incorrect.

6.
Elife ; 72018 12 06.
Article En | MEDLINE | ID: mdl-30520725

Interferon (IFN) inhibits HIV replication by inducing antiviral effectors. To comprehensively identify IFN-induced HIV restriction factors, we assembled a CRISPR sgRNA library of Interferon Stimulated Genes (ISGs) into a modified lentiviral vector that allows for packaging of sgRNA-encoding genomes in trans into budding HIV-1 particles. We observed that knockout of Zinc Antiviral Protein (ZAP) improved the performance of the screen due to ZAP-mediated inhibition of the vector. A small panel of IFN-induced HIV restriction factors, including MxB, IFITM1, Tetherin/BST2 and TRIM5alpha together explain the inhibitory effects of IFN on the CXCR4-tropic HIV-1 strain, HIV-1LAI, in THP-1 cells. A second screen with a CCR5-tropic primary strain, HIV-1Q23.BG505, described an overlapping, but non-identical, panel of restriction factors. Further, this screen also identifies HIV dependency factors. The ability of IFN-induced restriction factors to inhibit HIV strains to replicate in human cells suggests that these human restriction factors are incompletely antagonized. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Epithelial Cells/immunology , Gene Editing/methods , HIV-1/genetics , Host-Pathogen Interactions , Nuclear Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antiviral Restriction Factors , CRISPR-Cas Systems , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/virology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/immunology , HEK293 Cells , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Humans , Interferon-alpha/pharmacology , Lentivirus/genetics , Lentivirus/metabolism , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/immunology , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/immunology , RNA-Binding Proteins , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Repressor Proteins , Signal Transduction , THP-1 Cells , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Viral Tropism/genetics , Virus Assembly/drug effects , Virus Replication/drug effects
7.
Genome Med ; 10(1): 17, 2018 02 27.
Article En | MEDLINE | ID: mdl-29486792

BACKGROUND: Use of aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) has been shown to protect against tetraploidy, aneuploidy, and chromosomal alterations in the metaplastic condition Barrett's esophagus (BE) and to lower the incidence and mortality of esophageal adenocarcinoma (EA). The esophagus is exposed to both intrinsic and extrinsic mutagens resulting from gastric reflux, chronic inflammation, and exposure to environmental carcinogens such as those found in cigarettes. Here we test the hypothesis that NSAID use inhibits accumulation of point mutations/indels during somatic genomic evolution in BE. METHODS: Whole exome sequences were generated from 82 purified epithelial biopsies and paired blood samples from a cross-sectional study of 41 NSAID users and 41 non-users matched by sex, age, smoking, and continuous time using or not using NSAIDs. RESULTS: NSAID use reduced overall frequency of point mutations across the spectrum of mutation types, lowered the frequency of mutations even when adjusted for both TP53 mutation and smoking status, and decreased the prevalence of clones with high variant allele frequency. Never smokers who consistently used NSAIDs had fewer point mutations in signature 17, which is commonly found in EA. NSAID users had, on average, a 50% reduction in functional gene mutations in nine cancer-associated pathways and also had less diversity in pathway mutational burden compared to non-users. CONCLUSIONS: These results indicate NSAID use functions to limit overall mutations on which selection can act and supports a model in which specific mutant cell populations survive or expand better in the absence of NSAIDs.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Barrett Esophagus/drug therapy , Barrett Esophagus/genetics , Exome/genetics , Mutation/genetics , DNA Copy Number Variations/genetics , Gene Frequency/genetics , Humans , Loss of Heterozygosity , Mutagenesis/genetics
8.
Oncotarget ; 8(30): 48545-48562, 2017 Jul 25.
Article En | MEDLINE | ID: mdl-28596487

Zinc finger domain genes comprise ~3% of the human genome, yet many of their functions remain unknown. Here we investigated roles for the vertebrate-specific BTB domain zinc finger gene ZNF131 in the context of human brain tumors. We report that ZNF131 is broadly required for Glioblastoma stem-like cell (GSC) viability, but dispensable for neural progenitor cell (NPC) viability. Examination of gene expression changes after ZNF131 knockdown (kd) revealed that ZNF131 activity notably promotes expression of Joubert Syndrome ciliopathy genes, including KIF7, NPHP1, and TMEM237, as well as HAUS5, a component of Augmin/HAUS complex that facilitates microtubule nucleation along the mitotic spindle. Of these genes only kd of HAUS5 displayed GSC-specific viability loss. Critically, HAUS5 ectopic expression was sufficient to suppress viability defects of ZNF131 kd cells. Moreover, ZNF131 and HAUS5 kd phenocopied each other in GSCs, each causing: mitotic arrest, centrosome fragmentation, loss of Augmin/HAUS complex on the mitotic spindle, and loss of GSC self-renewal and tumor formation capacity. In control NPCs, we observed centrosome fragmentation and lethality only when HAUS5 kd was combined with kd of HAUS2 or HAUS4, demonstrating that the complex is essential in NPCs, but that GSCs have heightened requirement. Our results suggest that GSCs differentially rely on ZNF131-dependent expression of HAUS5 as well as the Augmin/HAUS complex activity to maintain the integrity of centrosome function and viability.


Brain Neoplasms/genetics , Centrosome/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Brain Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Binding , Spindle Apparatus/metabolism , Transcription Factors/metabolism
9.
Leuk Res ; 55: 23-32, 2017 04.
Article En | MEDLINE | ID: mdl-28119225

Dexamethasone (dex) induces apoptosis in multiple myeloma (MM) cells and is a frontline treatment for this disease. However resistance to dex remains a major challenge and novel treatment approaches are needed. We hypothesized that dex utilizes translational pathways to promote apoptosis in MM and that specific targeting of these pathways could overcome dex-resistance. Global unbiased profiling of mRNA translational profiles in MM cells treated with or without dex revealed that dex significantly repressed eIF2 signaling, an important pathway for regulating ternary complex formation and protein synthesis. We demonstrate that dex induces the phosphorylation of eIF2α resulting in the translational upregulation of ATF4, a known eIF2 regulated mRNA. Pharmacologic induction of eIF2α phosphorylation via activation of the heme-regulated eIF2α kinase (HRI) induced apoptosis in MM cell lines and in primary MM cells from patients with dex-resistant disease. In addition, co-culture with marrow stroma failed to protect MM cells from apoptosis induced by targeting the eIF2 pathway. Combination therapy with rapamycin, an mTOR inhibitor, and BTdCPU, an activator of HRI, demonstrated additive effects on apoptosis in dex-resistant cells. Thus, specific activation of the eIF2α kinase HRI is a novel therapeutic target in MM that can augment current treatment strategies.


Molecular Targeted Therapy/methods , Multiple Myeloma/drug therapy , eIF-2 Kinase/metabolism , Apoptosis/drug effects , Dexamethasone/pharmacology , Drug Resistance, Neoplasm , Humans , Phosphorylation , Protein Biosynthesis , Tumor Cells, Cultured , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/drug effects
10.
Immunity ; 45(2): 389-401, 2016 08 16.
Article En | MEDLINE | ID: mdl-27521269

CD8(+) T cells recognizing tumor-specific antigens are detected in cancer patients but are dysfunctional. Here we developed a tamoxifen-inducible liver cancer mouse model with a defined oncogenic driver antigen (SV40 large T-antigen) to follow the activation and differentiation of naive tumor-specific CD8(+) T (TST) cells after tumor initiation. Early during the pre-malignant phase of tumorigenesis, TST cells became dysfunctional, exhibiting phenotypic, functional, and transcriptional features similar to dysfunctional T cells isolated from late-stage human tumors. Thus, T cell dysfunction seen in advanced human cancers may already be established early during tumorigenesis. Although the TST cell dysfunctional state was initially therapeutically reversible, it ultimately evolved into a fixed state. Persistent antigen exposure rather than factors associated with the tumor microenvironment drove dysfunction. Moreover, the TST cell differentiation and dysfunction program exhibited features distinct from T cell exhaustion in chronic infections. Strategies to overcome this antigen-driven, cell-intrinsic dysfunction may be required to improve cancer immunotherapy.


Antigens, Polyomavirus Transforming/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Liver Neoplasms/immunology , Animals , Carcinogenesis , Cell Differentiation , Cells, Cultured , Cellular Senescence , Disease Models, Animal , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/therapy , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen , Tumor Microenvironment
11.
Mol Cancer Res ; 14(4): 374-84, 2016 Apr.
Article En | MEDLINE | ID: mdl-26753621

UNLABELLED: Prostate cancer-associated stroma (CAS) plays an active role in malignant transformation, tumor progression, and metastasis. Molecular analyses of CAS have demonstrated significant changes in gene expression; however, conflicting evidence exists on whether genomic alterations in benign cells comprising the tumor microenvironment (TME) underlie gene expression changes and oncogenic phenotypes. This study evaluates the nuclear and mitochondrial DNA integrity of prostate carcinoma cells, CAS, matched benign epithelium and benign epithelium-associated stroma by whole-genome copy-number analyses, targeted sequencing of TP53, and FISH. Array comparative genomic hybridization (aCGH) of CAS revealed a copy-neutral diploid genome with only rare and small somatic copy-number aberrations (SCNA). In contrast, several expected recurrent SCNAs were evident in the adjacent prostate carcinoma cells, including gains at 3q, 7p, and 8q, and losses at 8p and 10q. No somatic TP53 mutations were observed in CAS. Mitochondrial DNA (mtDNA) extracted from carcinoma cells and stroma identified 23 somatic mtDNA mutations in neoplastic epithelial cells, but only one mutation in stroma. Finally, genomic analyses identified no SCNAs, LOH, or copy-neutral LOH in cultured cancer-associated fibroblasts, which are known to promote prostate cancer progression in vivo IMPLICATIONS: The gene expression changes observed in prostate cancer-adjacent stroma and the attendant contribution of the stroma to the development and progression of prostate cancer are not due to frequent or recurrent genomic alterations in the TME.


Chromosome Aberrations , Chromosomes, Human/genetics , DNA, Mitochondrial/genetics , Prostatic Neoplasms/genetics , Comparative Genomic Hybridization , Gene Expression Regulation, Neoplastic , Humans , Male , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics
12.
Nat Biotechnol ; 33(6): 646-55, 2015 Jun.
Article En | MEDLINE | ID: mdl-25798938

Chromosomal deletions associated with human diseases, such as cancer, are common, but synteny issues complicate modeling of these deletions in mice. We use cellular reprogramming and genome engineering to functionally dissect the loss of chromosome 7q (del(7q)), a somatic cytogenetic abnormality present in myelodysplastic syndromes (MDS). We derive del(7q)- and isogenic karyotypically normal induced pluripotent stem cells (iPSCs) from hematopoietic cells of MDS patients and show that the del(7q) iPSCs recapitulate disease-associated phenotypes, including impaired hematopoietic differentiation. These disease phenotypes are rescued by spontaneous dosage correction and can be reproduced in karyotypically normal cells by engineering hemizygosity of defined chr7q segments in a 20-Mb region. We use a phenotype-rescue screen to identify candidate haploinsufficient genes that might mediate the del(7q)- hematopoietic defect. Our approach highlights the utility of human iPSCs both for functional mapping of disease-associated large-scale chromosomal deletions and for discovery of haploinsufficient genes.


Chromosome Deletion , Genetic Engineering , Induced Pluripotent Stem Cells/cytology , Myelodysplastic Syndromes/genetics , Animals , Chromosomes, Human, Pair 7/genetics , Humans , Karyotyping , Mice , Myelodysplastic Syndromes/therapy
13.
Curr Biol ; 25(6): 804-810, 2015 Mar 16.
Article En | MEDLINE | ID: mdl-25754639

The cytoplasmic functions of Wiskott-Aldrich syndrome family (WAS) proteins are well established and include roles in cytoskeleton reorganization and membrane-cytoskeletal interactions important for membrane/vesicle trafficking, morphogenesis, immune response, and signal transduction. Misregulation of these proteins is associated with immune deficiency and metastasis [1-4]. Cytoplasmic WAS proteins act as effectors of Rho family GTPases and polymerize branched actin through the Arp2/3 complex [1, 5]. Previously, we identified Drosophila washout (wash) as a new member of the WAS family with essential cytoplasmic roles in early development [6, 7]. Studies in mammalian cells and Dictyostelium suggest that WASH functions primarily in a multiprotein complex that regulates endosome shape and trafficking in an Arp2/3-dependent manner [8-11]. However, roles for classically cytoplasmic proteins in the nucleus are beginning to emerge, in particular, as participants in the regulation of gene expression [12, 13]. Here, we show that Drosophila Wash is present in the nucleus, where it plays a key role in global nuclear organization. wash mutant and knockdown nuclei disrupt subnuclear structures/organelles and exhibit the abnormal wrinkled morphology reminiscent of those observed in diverse laminopathies [14-16]. We find that nuclear Wash interacts with B-type Lamin (Lamin Dm0), and, like Lamin, Wash associates with constitutive heterochromatin. Wash knockdown increases chromatin accessibility of repressive compartments and results in a global redistribution of repressive histone modifications. Thus, our results reveal a novel role for Wash in modulating nucleus morphology and in the organization of both chromatin and non-chromatin nuclear sub-structures.


Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Lamins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/ultrastructure , Female , Gene Knockdown Techniques , Genes, Insect , Heterochromatin/genetics , Heterochromatin/metabolism , Lamins/genetics , Male , Mutation , Vesicular Transport Proteins/genetics
14.
Nat Genet ; 47(2): 180-5, 2015 Feb.
Article En | MEDLINE | ID: mdl-25581430

We report germline missense mutations in ETV6 segregating with the dominant transmission of thrombocytopenia and hematologic malignancy in three unrelated kindreds, defining a new hereditary syndrome featuring thrombocytopenia with susceptibility to diverse hematologic neoplasms. Two variants, p.Arg369Gln and p.Arg399Cys, reside in the highly conserved ETS DNA-binding domain. The third variant, p.Pro214Leu, lies within the internal linker domain, which regulates DNA binding. These three amino acid sites correspond to hotspots for recurrent somatic mutation in malignancies. Functional studies show that the mutations abrogate DNA binding, alter subcellular localization, decrease transcriptional repression in a dominant-negative fashion and impair hematopoiesis. These familial genetic studies identify a central role for ETV6 in hematopoiesis and malignant transformation. The identification of germline predisposition to cytopenias and cancer informs the diagnosis and medical management of at-risk individuals.


Hematologic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombocytopenia/genetics , Cell Proliferation , Exons/genetics , Female , Genes, Reporter , Germ-Line Mutation , HeLa Cells , Humans , Male , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Pedigree , Protein Structure, Tertiary , Recombinant Proteins , Sequence Analysis, RNA , ETS Translocation Variant 6 Protein
15.
J Immunol ; 194(4): 1677-85, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25582857

All aerobic cells and organisms must synthesize heme from the amino acid glycine and the tricarboxylic acid cycle intermediate succinyl CoA for incorporation into hemoproteins, such as the cytochromes needed for oxidative phosphorylation. Most studies on heme regulation have been done in erythroid cells or hepatocytes; however, much less is known about heme metabolism in other cell types. The feline leukemia virus subgroup C receptor (FLVCR) is a 12-transmembrane domain surface protein that exports heme from cells, and it was shown to be required for erythroid development. In this article, we show that deletion of Flvcr in murine hematopoietic precursors caused a complete block in αß T cell development at the CD4(+)CD8(+) double-positive stage, although other lymphoid lineages were not affected. Moreover, FLVCR was required for the proliferation and survival of peripheral CD4(+) and CD8(+) T cells. These studies identify a novel and unexpected role for FLVCR, a major facilitator superfamily metabolite transporter, in T cell development and suggest that heme metabolism is particularly important in the T lineage.


Cell Differentiation/immunology , Heme/immunology , Membrane Transport Proteins/immunology , Receptors, Virus/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Cell Separation , Cell Survival/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 9(10): e108942, 2014.
Article En | MEDLINE | ID: mdl-25291178

The stimulatory NKG2D receptor on lymphocytes promotes tumor immune surveillance by targeting ligands selectively induced on cancer cells. Progressing tumors counteract by employing tactics to disable lymphocyte NKG2D. This negative dynamic is escalated as some human cancer cells co-opt expression of NKG2D, thereby complementing the presence of its ligands for autonomous stimulation of oncogenic signaling. Clinical association data imply relationships between cancer cell NKG2D and metastatic disease. Here we show that NKG2D promotes cancer cell plasticity by induction of phenotypic, molecular, and functional signatures diagnostic of the epithelial-mesenchymal transition, and of stem-like traits via induction of Sox9, a key transcriptional regulator of breast stem cell maintenance. These findings obtained with model breast tumor lines and xenotransplants were recapitulated by ex vivo cancer cells from primary invasive breast carcinomas. Thus, NKG2D may have the capacity to drive high malignancy traits underlying metastatic disease.


Gene Expression , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Heterografts , Humans , Mice , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
17.
Stem Cells Dev ; 23(7): 729-40, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24131213

Marrow stromal cells constitute a heterogeneous population of cells, typically isolated after expansion in culture. In vivo, stromal cells often exist in close proximity or in direct contact with monocyte-derived macrophages, yet their interaction with monocytes is largely unexplored. In this report, isolated CD146(+) and CD146(-) stromal cells, as well as immortalized cell lines representative of each (designated HS27a and HS5, respectively), were shown by global DNase I hypersensitive site mapping and principal coordinate analysis to have a lineage association with marrow fibroblasts. Gene expression profiles generated for the CD146(+) and CD146(-) cell lines indicate significant differences in their respective transcriptomes, which translates into differences in secreted factors. Consequently, the conditioned media (CM) from these two populations induce different fates in peripheral blood monocytes. Monocytes incubated in CD146(+) CM acquire a tissue macrophage phenotype, whereas monocytes incubated in CM from CD146(-) cells express markers associated with pre-dendritic cells. Importantly, when CD14(+) monocytes are cultured in contact with the CD146(+) cells, the combined cell populations, assayed as a unit, show increased levels of transcripts associated with organismal development and hematopoietic regulation. In contrast, the gene expression profile from cocultures of monocytes and CD146(-) cells does not differ from that obtained when monocytes are cultured with CD146(-) CM. These in vitro results show that the CD146(+) marrow stromal cells together with monocytes increase the expression of genes relevant to hematopoietic regulation. In vivo relevance of these data is suggested by immunohistochemistry of marrow biopsies showing juxtaposed CD146(+) cells and CD68(+) cells associated with these upregulated proteins.


Adult Stem Cells/cytology , Bone Marrow Cells/cytology , Fibroblasts/cytology , Mesenchymal Stem Cells/cytology , Monocytes/cytology , Adult , Adult Stem Cells/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/metabolism , Bone Marrow Cells/metabolism , CD146 Antigen/genetics , CD146 Antigen/metabolism , Cell Differentiation , Cell Line, Transformed , Cell Lineage/physiology , Chromosome Mapping , Cytokines/genetics , Cytokines/metabolism , Deoxyribonuclease I/chemistry , Fibroblasts/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling , Humans , Immunophenotyping , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Monocytes/metabolism
18.
AIDS Res Hum Retroviruses ; 29(10): 1321-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-23815575

The potential role of conventional and regulatory T cells (Tregs) in protection from HIV-1 infection remains unclear. To address this question, we analyzed samples from 129 HIV-1-exposed seronegative individuals (HESN) from an HIV-1-serodiscordant couples cohort. To assess the presence of HIV-specific T cell responses and Treg function, we measured the proliferation of T cells in response to HIV-1 peptide pools in peripheral blood mononuclear cells (PBMCs) and PBMCs depleted of Tregs. We identified HIV-specific CD4(+) and CD8(+) T cell responses and, surprisingly, the overall CD4(+) and CD8(+) T cell response rate was not increased when Tregs were removed from cell preparations. Of the 20 individuals that had HIV-1-specific CD4(+) T cell responses, only eight had Tregs that could suppress this proliferation. When compared with individuals whose Tregs could suppress HIV-1-specific CD4(+) T cell proliferation, individuals with Tregs unable to suppress showed a trend toward increased T cell activation and Treg frequency and a significant increase in HIV-1-specific production of microphage inflammatory protein-1ß (MIP-1ß) by CD4(+) T cells, autocrine production of which has been shown to be protective in terms of HIV-1 infection of CD4(+) T cells.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV-1/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Cell Proliferation , Chemokine CCL4/metabolism , Female , Humans , Leukocytes, Mononuclear/immunology , Male
19.
Cell ; 151(6): 1214-28, 2012 Dec 07.
Article En | MEDLINE | ID: mdl-23177352

Developmental gene expression results from the orchestrated interplay between genetic and epigenetic mechanisms. Here, we describe upSET, a transcriptional regulator encoding a SET domain-containing protein recruited to active and inducible genes in Drosophila. However, unlike other Drosophila SET proteins associated with gene transcription, UpSET is part of an Rpd3/Sin3-containing complex that restricts chromatin accessibility and histone acetylation to promoter regions. In the absence of UpSET, active chromatin marks and chromatin accessibility increase and spread to genic and flanking regions due to destabilization of the histone deacetylase complex. Consistent with this, transcriptional noise increases, as manifest by activation of repetitive elements and off-target genes. Interestingly, upSET mutant flies are female sterile due to upregulation of key components of Notch signaling during oogenesis. Thus UpSET defines a class of metazoan transcriptional regulators required to fine tune transcription by preventing the spread of active chromatin.


Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Histone Deacetylases/metabolism , Promoter Regions, Genetic , Acetylation , Animals , Chromatin , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Female , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Mutation
20.
Mol Cell Biol ; 32(11): 2160-7, 2012 Jun.
Article En | MEDLINE | ID: mdl-22473991

Colorectal cancer (CRC) remains a major cause of cancer mortality worldwide. Murine models have yielded critical insights into CRC pathogenesis, but they often fail to recapitulate advanced-disease phenotypes, notably metastasis and chromosomal instability (CIN). New models are thus needed to understand disease progression and to develop therapies. We sought to model advanced CRC by inactivating two tumor suppressors that are mutated in human CRCs, the Fbw7 ubiquitin ligase and p53. Here we report that Fbw7 deletion alters differentiation and proliferation in the gut epithelium and stabilizes oncogenic Fbw7 substrates, such as cyclin E and Myc. However, Fbw7 deletion does not cause tumorigenesis in the gut. In contrast, codeletion of both Fbw7 and p53 causes highly penetrant, aggressive, and metastatic adenocarcinomas, and allografts derived from these tumors form highly malignant adenocarcinomas. In vitro evidence indicates that Fbw7 ablation promotes genetic instability that is suppressed by p53, and we show that most Fbw7⁻/⁻; p53⁻/⁻ carcinomas exhibit a CIN⁺ phenotype. We conclude that Fbw7 and p53 synergistically suppress adenocarcinomas that mimic advanced human CRC with respect to histopathology, metastasis, and CIN. This model thus represents a novel tool for studies of advanced CRC as well as carcinogenesis associated with ubiquitin pathway mutations.


Adenocarcinoma , Cell Cycle Proteins/metabolism , Colorectal Neoplasms , Disease Models, Animal , F-Box Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cell Transformation, Neoplastic , Chromosomal Instability , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
...