Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Genet ; 14: 1192799, 2023.
Article in English | MEDLINE | ID: mdl-37229187

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous and deadly disease characterized by uncontrolled expansion of malignant blasts. Altered metabolism and dysregulated microRNA (miRNA) expression profiles are both characteristic of AML. However, there is a paucity of studies exploring how changes in the metabolic state of the leukemic cells regulate miRNA expression leading to altered cellular behavior. Here, we blocked pyruvate entry into mitochondria by deleting the Mitochondria Pyruvate Carrier (MPC1) gene in human AML cell lines, which decreased Oxidative Phosphorylation (OXPHOS). This metabolic shift also led to increased expression of miR-1 in the human AML cell lines tested. AML patient sample datasets showed that higher miR-1 expression correlates with reduced survival. Transcriptional and metabolic profiling of miR-1 overexpressing AML cells revealed that miR-1 increased OXPHOS, along with key metabolites that fuel the TCA cycle such as glutamine and fumaric acid. Inhibition of glutaminolysis decreased OXPHOS in miR-1 overexpressing MV4-11 cells, highlighting that miR-1 promotes OXPHOS through glutaminolysis. Finally, overexpression of miR-1 in AML cells exacerbated disease in a mouse xenograft model. Together, our work expands current knowledge within the field by uncovering novel connections between AML cell metabolism and miRNA expression that facilitates disease progression. Further, our work points to miR-1 as a potential new therapeutic target that may be used to disrupt AML cell metabolism and thus pathogenesis in the clinic.

2.
Virology ; 578: 154-162, 2023 01.
Article in English | MEDLINE | ID: mdl-36577173

ABSTRACT

A primary function of HIV-1 Nef is the enhancement of viral infectivity and replication. Whether counteraction of the antiretroviral proteins SERINC3 and SERINC5 is the cause of this positive influence on viral growth-rate and infectivity remains unclear. Here, we utilized CRISPR/Cas9 to knockout SERINC3 and SERINC5 in a leukemic CD4-positive T cell line (CEM) that displays nef-related infectivity and growth-rate phenotypes. Viral replication was attenuated in CEM cells infected with HIV-1 lacking Nef (HIV-1ΔNef). This attenuated growth-rate phenotype was observed regardless of whether the coding regions of the serinc3 or serinc5 genes were intact. Moreover, knockout of serinc5 alone or of both serinc5 and serinc3 together failed to restore the infectivity of HIV1ΔNef virions produced from infected CEM cells. Our results corroborate a similar study using another T-lymphoid cell line (MOLT-3) and indicate that the antagonism of SERINC3 and SERINC5 does not fully explain the virology of HIV-1 lacking Nef.


Subject(s)
HIV-1 , Membrane Proteins , CD4-Positive T-Lymphocytes/metabolism , HIV-1/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism , Virus Replication/genetics
3.
J Biol Chem ; 295(32): 11002-11020, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32518166

ABSTRACT

Mitochondrial iron import is essential for iron-sulfur cluster formation and heme biosynthesis. Two nuclear-encoded vertebrate mitochondrial high-affinity iron importers, mitoferrin1 (Mfrn1) and Mfrn2, have been identified in mammals. In mice, the gene encoding Mfrn1, solute carrier family 25 member 37 (Slc25a37), is highly expressed in sites of erythropoiesis, and whole-body Slc25a37 deletion leads to lethality. Here, we report that mice with a deletion of Slc25a28 (encoding Mfrn2) are born at expected Mendelian ratios, but show decreased male fertility due to reduced sperm numbers and sperm motility. Mfrn2-/- mice placed on a low-iron diet exhibited reduced mitochondrial manganese, cobalt, and zinc levels, but not reduced iron. Hepatocyte-specific loss of Slc25a37 (encoding Mfrn1) in Mfrn2-/- mice did not affect animal viability, but resulted in a 40% reduction in mitochondrial iron and reduced levels of oxidative phosphorylation proteins. Placing animals on a low-iron diet exaggerated the reduction in mitochondrial iron observed in liver-specific Mfrn1/2-knockout animals. Mfrn1-/-/Mfrn2-/- bone marrow-derived macrophages or skin fibroblasts in vitro were unable to proliferate, and overexpression of Mfrn1-GFP or Mfrn2-GFP prevented this proliferation defect. Loss of both mitoferrins in hepatocytes dramatically reduced regeneration in the adult mouse liver, further supporting the notion that both mitoferrins transport iron and that their absence limits proliferative capacity of mammalian cells. We conclude that Mfrn1 and Mfrn2 contribute to mitochondrial iron homeostasis and are required for high-affinity iron import during active proliferation of mammalian cells.


Subject(s)
Cation Transport Proteins/physiology , Cell Proliferation/physiology , Liver Regeneration/physiology , Membrane Transport Proteins/physiology , Animals , Homeostasis , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism
4.
Cell Rep ; 30(9): 2889-2899.e6, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130894

ABSTRACT

Metabolic pathways regulate T cell development and function, but many remain understudied. Recently, the mitochondrial pyruvate carrier (MPC) was identified as the transporter that mediates pyruvate entry into mitochondria, promoting pyruvate oxidation. Here we find that deleting Mpc1, an obligate MPC subunit, in the hematopoietic system results in a specific reduction in peripheral αß T cell numbers. MPC1-deficient T cells have defective thymic development at the ß-selection, intermediate single positive (ISP)-to-double-positive (DP), and positive selection steps. We find that early thymocytes deficient in MPC1 display alterations to multiple pathways involved in T cell development. This results in preferred escape of more activated T cells. Finally, mice with hematopoietic deletion of Mpc1 are more susceptible to experimental autoimmune encephalomyelitis. Altogether, our study demonstrates that pyruvate oxidation by T cell precursors is necessary for optimal αß T cell development and that its deficiency results in reduced but activated peripheral T cell populations.


Subject(s)
Anion Transport Proteins/metabolism , Homeostasis , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , T-Lymphocytes/metabolism , Thymus Gland/growth & development , Thymus Gland/metabolism , Animals , Anion Transport Proteins/deficiency , Gene Deletion , Glycolysis , Hematopoiesis , Humans , Inflammation/pathology , Jurkat Cells , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/deficiency , Monocarboxylic Acid Transporters/deficiency , Oxidation-Reduction , Oxidative Phosphorylation , Pyruvic Acid/metabolism , Thymocytes/metabolism
5.
J Immunol ; 204(8): 2064-2075, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32161096

ABSTRACT

Aging-related chronic inflammation is a risk factor for many human disorders through incompletely understood mechanisms. Aged mice deficient in microRNA (miRNA/miR)-146a succumb to life-shortening chronic inflammation. In this study, we report that miR-155 in T cells contributes to shortened lifespan of miR-146a-/- mice. Using single-cell RNA sequencing and flow cytometry, we found that miR-155 promotes the activation of effector T cell populations, including T follicular helper cells, and increases germinal center B cells and autoantibodies in mice aged over 15 months. Mechanistically, aerobic glycolysis genes are elevated in T cells during aging, and upon deletion of miR-146a, in a T cell miR-155-dependent manner. Finally, skewing T cell metabolism toward aerobic glycolysis by deleting mitochondrial pyruvate carrier recapitulates age-dependent T cell phenotypes observed in miR-146a-/- mice, revealing the sufficiency of metabolic reprogramming to influence immune cell functions during aging. Altogether, these data indicate that T cell-specific miRNAs play pivotal roles in regulating lifespan through their influences on inflammaging.


Subject(s)
Disease Models, Animal , Inflammation/genetics , Longevity/genetics , MicroRNAs/genetics , T-Lymphocytes/metabolism , Age Factors , Animals , Female , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
PLoS Genet ; 15(2): e1007970, 2019 02.
Article in English | MEDLINE | ID: mdl-30768595

ABSTRACT

Identifying regulatory mechanisms that influence inflammation in metabolic tissues is critical for developing novel metabolic disease treatments. Here, we investigated the role of microRNA-146a (miR-146a) during diet-induced obesity in mice. miR-146a is reduced in obese and type 2 diabetic patients and our results reveal that miR-146a-/- mice fed a high-fat diet (HFD) have exaggerated weight gain, increased adiposity, hepatosteatosis, and dysregulated blood glucose levels compared to wild-type controls. Pro-inflammatory genes and NF-κB activation increase in miR-146a-/- mice, indicating a role for this miRNA in regulating inflammatory pathways. RNA-sequencing of adipose tissue macrophages demonstrated a role for miR-146a in regulating both inflammation and cellular metabolism, including the mTOR pathway, during obesity. Further, we demonstrate that miR-146a regulates inflammation, cellular respiration and glycolysis in macrophages through a mechanism involving its direct target Traf6. Finally, we found that administration of rapamycin, an inhibitor of mTOR, was able to rescue the obesity phenotype in miR-146a-/- mice. Altogether, our study provides evidence that miR-146a represses inflammation and diet-induced obesity and regulates metabolic processes at the cellular and organismal levels, demonstrating how the combination of diet and miRNA genetics influences obesity and diabetic phenotypes.


Subject(s)
Inflammation/prevention & control , Metabolic Diseases/prevention & control , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Expression , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Inflammation/genetics , Inflammation/metabolism , Insulin/blood , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Macrophages/metabolism , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , NF-kappa B/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/prevention & control , Proto-Oncogene Proteins c-akt/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Weight Gain/drug effects , Weight Gain/genetics
7.
J Immunol ; 199(10): 3559-3570, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28978688

ABSTRACT

Extracellular vesicles, including exosomes, have recently been implicated as novel mediators of immune cell communication in mammals. However, roles for endogenously produced exosomes in regulating immune cell functions in vivo are just beginning to be identified. In this article, we demonstrate that Rab27a and Rab27b double-knockout (Rab27DKO) mice that are deficient in exosome secretion have a chronic, low-grade inflammatory phenotype characterized by elevated inflammatory cytokines and myeloproliferation. Upon further investigation, we found that some of these phenotypes could be complemented by wild-type (WT) hematopoietic cells or administration of exosomes produced by GM-CSF-expanded bone marrow cells. In addition, chronically inflamed Rab27DKO mice had a blunted response to bacterial LPS, resembling endotoxin tolerance. This defect was rescued by bone marrow exosomes from WT, but not miR-155-/-, cells, suggesting that uptake of miR-155-containing exosomes is important for a proper LPS response. Further, we found that SHIP1 and IRAK-M, direct targets of miR-155 that are known negative regulators of the LPS response, were elevated in Rab27DKO mice and decreased after treatment with WT, but not miR-155-/-, exosomes. Together, our study finds that Rab27-dependent exosome production contributes to homeostasis within the hematopoietic system and appropriate responsiveness to inflammatory stimuli.


Subject(s)
Exosomes/metabolism , Inflammation/immunology , MicroRNAs/genetics , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins/metabolism , Acute Disease , Animals , Cell Proliferation , Cells, Cultured , Chronic Disease , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immune Tolerance , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/pathology , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins/genetics
8.
J Biol Chem ; 292(45): 18530-18541, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28912267

ABSTRACT

MicroRNA-155 (miR-155) regulates antitumor immune responses. However, its specific functions within distinct immune cell types have not been delineated in conditional KO mouse models. In this study, we investigated the role of miR-155 specifically within T cells during the immune response to syngeneic tumors. We found that miR-155 expression within T cells is required to limit syngeneic tumor growth and promote IFNγ production by T cells within the tumor microenvironment. Consequently, we found that miR-155 expression by T cells is necessary for proper tumor-associated macrophage expression of IFNγ-inducible genes. We also found that immune checkpoint-blocking (ICB) antibodies against programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) restored antitumor immunity in miR-155 T cell-conditional KO mice. We noted that these ICB antibodies rescued the levels of IFNγ-expressing T cells, expression of multiple activation and effector genes expressed by tumor-infiltrating CD8+ and CD4+ T cells, and tumor-associated macrophage activation. Moreover, the ICB approach partially restored expression of several derepressed miR-155 targets in tumor-infiltrating, miR-155-deficient CD8+ T cells, suggesting that miR-155 and ICB regulate overlapping pathways to promote antitumor immunity. Taken together, our findings highlight the multifaceted role of miR-155 in T cells, in which it promotes antitumor immunity. These results suggest that the augmentation of miR-155 expression could be used to improve anticancer immunotherapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma/drug therapy , MicroRNAs/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Blocking/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Crosses, Genetic , Immunologic Surveillance/drug effects , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
9.
J Biol Chem ; 292(39): 16284-16299, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28808058

ABSTRACT

ATP-binding cassette subfamily B member 10 (Abcb10) is a mitochondrial ATP-binding cassette (ABC) transporter that complexes with mitoferrin1 and ferrochelatase to enhance heme biosynthesis in developing red blood cells. Reductions in Abcb10 levels have been shown to reduce mitoferrin1 protein levels and iron import into mitochondria, resulting in reduced heme biosynthesis. As an ABC transporter, Abcb10 binds and hydrolyzes ATP, but its transported substrate is unknown. Here, we determined that decreases in Abcb10 did not result in protoporphyrin IX accumulation in morphant-treated zebrafish embryos or in differentiated Abcb10-specific shRNA murine Friend erythroleukemia (MEL) cells in which Abcb10 was specifically silenced with shRNA. We also found that the ATPase activity of Abcb10 is necessary for hemoglobinization in MEL cells, suggesting that the substrate transported by Abcb10 is important in mediating increased heme biosynthesis during erythroid development. Inhibition of 5-aminolevulinic acid dehydratase (EC 4.2.1.24) with succinylacetone resulted in both 5-aminolevulinic acid (ALA) accumulation in control and Abcb10-specific shRNA MEL cells, demonstrating that reductions in Abcb10 do not affect ALA export from mitochondria and indicating that Abcb10 does not transport ALA. Abcb10 silencing resulted in an alteration in the heme biosynthesis transcriptional profile due to repression by the transcriptional regulator Bach1, which could be partially rescued by overexpression of Alas2 or Gata1, providing a mechanistic explanation for why Abcb10 shRNA MEL cells exhibit reduced hemoglobinization. In conclusion, our findings rule out that Abcb10 transports ALA and indicate that Abcb10's ATP-hydrolysis activity is critical for hemoglobinization and that the substrate transported by Abcb10 provides a signal that optimizes hemoglobinization.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Heme/biosynthesis , Zebrafish Proteins/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Motifs , Amino Acid Substitution , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/genetics , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Fanconi Anemia Complementation Group Proteins , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Microinjections , Morpholinos/metabolism , Mutation , RNA Interference , RNA, Small Interfering , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
10.
Blood ; 130(11): 1290-1301, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28751524

ABSTRACT

Acute myeloid leukemia (AML) is a deadly hematologic malignancy characterized by the uncontrolled growth of immature myeloid cells. Over the past several decades, we have learned a tremendous amount regarding the genetic aberrations that govern disease development in AML. Among these are genes that encode noncoding RNAs, including the microRNA (miRNA) family. miRNAs are evolutionarily conserved small noncoding RNAs that display important physiological effects through their posttranscriptional regulation of messenger RNA targets. Over the past decade, studies have identified miRNAs as playing a role in nearly all aspects of AML disease development, including cellular proliferation, survival, and differentiation. These observations have led to the study of miRNAs as biomarkers of disease, and efforts to therapeutically manipulate miRNAs to improve disease outcome in AML are ongoing. Although much has been learned regarding the importance of miRNAs in AML disease initiation and progression, there are many unanswered questions and emerging facets of miRNA biology that add complexity to their roles in AML. Moving forward, answers to these questions will provide a greater level of understanding of miRNA biology and critical insights into the many translational applications for these small regulatory RNAs in AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , MicroRNAs/genetics , Translational Research, Biomedical , Animals , Exosomes/metabolism , Gene Expression Regulation, Leukemic , Humans , MicroRNAs/metabolism , Models, Biological
11.
Blood ; 129(23): 3074-3086, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28432220

ABSTRACT

FLT3-ITD+ acute myeloid leukemia (AML) accounts for ∼25% of all AML cases and is a subtype that carries a poor prognosis. microRNA-155 (miR-155) is specifically overexpressed in FLT3-ITD+ AML compared with FLT3 wild-type (FLT3-WT) AML and is critical for the growth of FLT3-ITD+ AML cells in vitro. However, miR-155's role in regulating FLT3-ITD-mediated disease in vivo remains unclear. In this study, we used a genetic mouse model to determine whether miR-155 influences the development of FLT3-ITD-induced myeloproliferative disease. Results indicate that miR-155 promotes FLT3-ITD-induced myeloid expansion in the bone marrow, spleen, and peripheral blood. Mechanistically, miR-155 increases proliferation of the hematopoietic stem and progenitor cell compartments by reducing the growth-inhibitory effects of the interferon (IFN) response, and this involves targeting of Cebpb. Consistent with our observations in mice, primary FLT3-ITD+ AML clinical samples have significantly higher miR-155 levels and a lower IFN response compared with FLT3-WT AML samples. Further, inhibition of miR-155 in FLT3-ITD+ AML cell lines using CRISPR/Cas9, or primary FLT3-ITD+ AML samples using locked nucleic acid antisense inhibitors, results in an elevated IFN response and reduces colony formation. Altogether, our data reveal that miR-155 collaborates with FLT3-ITD to promote myeloid cell expansion in vivo and that this involves a multitarget mechanism that includes repression of IFN signaling.


Subject(s)
Interferons/biosynthesis , MicroRNAs/genetics , Myeloproliferative Disorders/etiology , fms-Like Tyrosine Kinase 3/genetics , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , MicroRNAs/antagonists & inhibitors , Mutation , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Myelopoiesis/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Tumor Stem Cell Assay
12.
J Clin Invest ; 126(10): 3783-3798, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27599294

ABSTRACT

Neutrophil granulocytes, also called polymorphonuclear leukocytes (PMNs), extrude molecular lattices of decondensed chromatin studded with histones, granule enzymes, and antimicrobial peptides that are referred to as neutrophil extracellular traps (NETs). NETs capture and contain bacteria, viruses, and other pathogens. Nevertheless, experimental evidence indicates that NETs also cause inflammatory vascular and tissue damage, suggesting that identifying pathways that inhibit NET formation may have therapeutic implications. Here, we determined that neonatal NET-inhibitory factor (nNIF) is an inhibitor of NET formation in umbilical cord blood. In human neonatal and adult neutrophils, nNIF inhibits key terminal events in NET formation, including peptidyl arginine deiminase 4 (PAD4) activity, neutrophil nuclear histone citrullination, and nuclear decondensation. We also identified additional nNIF-related peptides (NRPs) that inhibit NET formation. nNIFs and NRPs blocked NET formation induced by pathogens, microbial toxins, and pharmacologic agonists in vitro and in mouse models of infection and systemic inflammation, and they improved mortality in murine models of systemic inflammation, which are associated with NET-induced collateral tissue injury. The identification of NRPs as neutrophil modulators that selectively interrupt NET generation at critical steps suggests their potential as therapeutic agents. Furthermore, our results indicate that nNIF may be an important regulator of NET formation in fetal and neonatal inflammation.


Subject(s)
Blood Proteins/physiology , Extracellular Traps/metabolism , Neutrophils/metabolism , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Fetal Blood/metabolism , Histones/metabolism , Humans , Infant, Newborn , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Neoplasm Proteins/physiology , Neutrophils/immunology , Protein Processing, Post-Translational
13.
PLoS One ; 11(4): e0153689, 2016.
Article in English | MEDLINE | ID: mdl-27081855

ABSTRACT

Mammalian microRNA expression is dysregulated in human cancer. However, the functional relevance of many microRNAs in the context of tumor biology remains unclear. Using CRISPR-Cas9 technology, we performed a global loss-of-function screen to simultaneously test the functions of individual microRNAs and protein-coding genes during the growth of a myeloid leukemia cell line. This approach identified evolutionarily conserved human microRNAs that suppress or promote cell growth, revealing that microRNAs are extensively integrated into the molecular networks that control tumor cell physiology. miR-155 was identified as a top microRNA candidate promoting cellular fitness, which we confirmed with two distinct miR-155-targeting CRISPR-Cas9 lentiviral constructs. Further, we performed anti-correlation functional profiling to predict relevant microRNA-tumor suppressor gene or microRNA-oncogene interactions in these cells. This analysis identified miR-150 targeting of p53, a connection that was experimentally validated. Taken together, our study describes a powerful genetic approach by which the function of individual microRNAs can be assessed on a global level, and its use will rapidly advance our understanding of how microRNAs contribute to human disease.


Subject(s)
CRISPR-Cas Systems , Genomics , Leukemia, Myeloid/pathology , MicroRNAs/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Conserved Sequence , Evolution, Molecular , Humans , Leukemia, Myeloid/genetics , Mice , Molecular Sequence Data , Tumor Suppressor Protein p53/genetics
14.
Oncotarget ; 6(30): 28556-72, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26456940

ABSTRACT

Host-microbial interactions within the mammalian intestines must be properly regulated in order to promote host health and limit disease. Because the microbiota provide constant immunological signals to intestinal tissues, a variety of regulatory mechanisms have evolved to ensure proper immune responses to maintain homeostasis. However, many of the genes that comprise these regulatory pathways, including immune-modulating microRNAs (miRNAs), have not yet been identified or studied in the context of intestinal homeostasis. Here, we investigated the role of microRNA-146a (miR-146a) in regulating intestinal immunity and barrier function and found that this miRNA is expressed in a variety of gut tissues in adult mice. By comparing intestinal gene expression in WT and miR-146a-/- mice, we demonstrate that miR-146a represses a subset of gut barrier and inflammatory genes all within a network of immune-related signaling pathways. We also found that miR-146a restricts the expansion of intestinal T cell populations, including Th17, Tregs, and Tfh cells. GC B cells, Tfh ICOS expression, and the production of luminal IgA were also reduced by miR-146a in the gut. Consistent with an enhanced intestinal barrier, we found that miR-146a-/- mice are resistant to DSS-induced colitis, a model of Ulcerative Colitis (UC), and this correlated with elevated colonic miR-146a expression in human UC patients. Taken together, our data describe a role for miR-146a in constraining intestinal barrier function, a process that alters gut homeostasis and enhances at least some forms of intestinal disease in mice.


Subject(s)
Colitis/immunology , Colon/immunology , Dextran Sulfate , Immunity, Mucosal , MicroRNAs/immunology , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/microbiology , Colitis/pathology , Colitis/prevention & control , Colon/metabolism , Colon/microbiology , Colon/pathology , Disease Models, Animal , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Genetic Predisposition to Disease , Host-Pathogen Interactions , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Phenotype , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors
15.
PLoS One ; 8(5): e63074, 2013.
Article in English | MEDLINE | ID: mdl-23667577

ABSTRACT

MicroRNAs (miRNAs) have quickly emerged as important regulators of mammalian physiology owing to their precise control over the expression of critical protein coding genes. Despite significant progress in our understanding of how miRNAs function in mice, there remains a fundamental need to be able to target and edit miRNA genes in the human genome. Here, we report a novel approach to disrupting human miRNA genes ex vivo using engineered TAL-effector (TALE) proteins to function as nucleases (TALENs) that specifically target and disrupt human miRNA genes. We demonstrate that functional TALEN pairs can be designed to enable disruption of miRNA seed regions, or removal of entire hairpin sequences, and use this approach to successfully target several physiologically relevant human miRNAs including miR-155*, miR-155, miR-146a and miR-125b. This technology will allow for a substantially improved capacity to study the regulation and function of miRNAs in human cells, and could be developed into a strategic means by which miRNAs can be targeted therapeutically during human disease.


Subject(s)
Endonucleases/metabolism , Genetic Engineering , MicroRNAs/genetics , Alleles , Animals , Base Sequence , Genetic Loci/genetics , Genome, Human/genetics , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Mutation Rate , Nucleic Acid Conformation , Sequence Deletion/genetics , Transfection
16.
Antimicrob Agents Chemother ; 55(12): 5753-60, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947398

ABSTRACT

Zinc pyrithione (ZPT) is an antimicrobial material with widespread use in antidandruff shampoos and antifouling paints. Despite decades of commercial use, there is little understanding of its antimicrobial mechanism of action. We used a combination of genome-wide approaches (yeast deletion mutants and microarrays) and traditional methods (gene constructs and atomic emission) to characterize the activity of ZPT against a model yeast, Saccharomyces cerevisiae. ZPT acts through an increase in cellular copper levels that leads to loss of activity of iron-sulfur cluster-containing proteins. ZPT was also found to mediate growth inhibition through an increase in copper in the scalp fungus Malassezia globosa. A model is presented in which pyrithione acts as a copper ionophore, enabling copper to enter cells and distribute across intracellular membranes. This is the first report of a metal-ligand complex that inhibits fungal growth by increasing the cellular level of a different metal.


Subject(s)
Antifungal Agents/pharmacology , Copper/metabolism , Iron-Sulfur Proteins/antagonists & inhibitors , Malassezia/drug effects , Organometallic Compounds/pharmacology , Pyridines/pharmacology , Saccharomyces cerevisiae/drug effects , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Humans , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Malassezia/genetics , Malassezia/growth & development , Oligonucleotide Array Sequence Analysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sequence Deletion
17.
Blood ; 117(20): 5494-502, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21310927

ABSTRACT

Mitoferrin1 is 1 of 2 homologous mitochondrial iron transporters and is required for mitochondrial iron delivery in developing erythroid cells. We show that total deletion of Mfrn1 in embryos leads to embryonic lethality. Selective deletion of Mfrn1 in adult hematopoietic tissues leads to severe anemia because of a deficit in erythroblast formation. Deletion of Mfrn1 in hepatocytes has no phenotype or biochemical effect under normal conditions. In the presence of increased porphyrin synthesis, however, deletion of Mfrn1 in hepatocytes results in a decreased ability to convert protoporphyrin IX into heme, leading to protoporphyria, cholestasis, and bridging cirrhosis. Our results show that the activity of mitoferrin1 is required to manage an increase in heme synthesis. The data also show that alterations in heme synthesis within hepatocytes can lead to protoporphyria and hepatotoxicity.


Subject(s)
Anemia/etiology , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Protoporphyria, Erythropoietic/etiology , Anemia/genetics , Animals , Base Sequence , DNA Primers/genetics , Embryo Loss/genetics , Female , Gene Targeting , Heme/biosynthesis , Hepatocytes/metabolism , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Pregnancy , Protoporphyria, Erythropoietic/genetics , Protoporphyrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...