Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cell Rep ; 36(3): 109430, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289356

ABSTRACT

While the intrinsic apoptosis pathway is thought to play a central role in shaping the B cell lineage, its precise role in mature B cell homeostasis remains elusive. Using mice in which mature B cells are unable to undergo apoptotic cell death, we show that apoptosis constrains follicular B (FoB) cell lifespan but plays no role in marginal zone B (MZB) cell homeostasis. In these mice, FoB cells accumulate abnormally. This intensifies intercellular competition for BAFF, resulting in a contraction of the MZB cell compartment, and reducing the growth, trafficking, and fitness of FoB cells. Diminished BAFF signaling dampens the non-canonical NF-κB pathway, undermining FoB cell growth despite the concurrent triggering of a protective p53 response. Thus, MZB and FoB cells exhibit a differential requirement for the intrinsic apoptosis pathway. Homeostatic apoptosis constrains the size of the FoB cell compartment, thereby preventing competition-induced FoB cell atrophy.


Subject(s)
Apoptosis , B-Lymphocytes/pathology , Homeostasis , Animals , Antibody Formation/immunology , Atrophy , B-Cell Activating Factor/metabolism , Cell Count , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Size , Cell Survival/genetics , Cellular Senescence/genetics , Gene Deletion , Gene Expression Regulation , Mice, Knockout , Sequence Analysis, RNA , Thymus Gland/immunology , Transcription Factors/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
3.
Curr Protoc ; 1(4): e79, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33836122

ABSTRACT

The generation of radiation chimeras allows researchers to substitute the hematopoietic system of a mouse with that of one or more donors. A suspension of hematopoietic stem cells (HSCs) is prepared from the bone marrow (BM) or the fetal liver (FL) of a donor mouse and adoptively transferred into an irradiated recipient. Within days, the donor's HSCs will engraft, and their progeny will quickly replace the blood cells of the recipient. This simple tool, together with the large availability of genetically modified mouse lines, can be harnessed to manipulate and study various aspects of blood cell biology in vivo. We present here protocols to generate three types of radiation chimera: (1) BM chimeras, which can assist in determining whether the origin of a genetically based phenotype is the hematopoietic or radio-resistant compartment and which are also conducive for studying the ecology of blood cells and for manipulating the environment hematopoietic cells live; (2) FL chimeras, which allow the study of hematopoietic systems from animals that carry genetic modifications incompatible with postnatal life; and (3) mixed BM chimeras, in which the hematopoietic system comprises blood cells of two different genotypes. Mixed BM chimeras can be used to identify genes that affect hematopoietic cell fitness and to establish whether secreted factors mediate a phenotype of interest. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Generation of bone marrow chimera Basic Protocol 2: Generation of fetal liver chimera Basic Protocol 3: Generation of mixed bone marrow chimera Support Protocol 1: Isolation of bone marrow cells Support Protocol 2: Cell counting by flow cytometry Support Protocol 3: Assessment of chimerism.


Subject(s)
Bone Marrow , Chimera , Animals , Hematopoietic Stem Cells , Liver , Mice , Radiation Chimera
4.
Nat Chem Biol ; 17(4): 428-437, 2021 04.
Article in English | MEDLINE | ID: mdl-33542533

ABSTRACT

Tryptophan C-mannosylation is an unusual co-translational protein modification performed by metazoans and apicomplexan protists. The prevalence and biological functions of this modification are poorly understood, with progress in the field hampered by a dearth of convenient tools for installing and detecting the modification. Here, we engineer a yeast system to produce a diverse array of proteins with and without tryptophan C-mannosylation and interrogate the modification's influence on protein stability and function. This system also enabled mutagenesis studies to identify residues of the glycosyltransferase and its protein substrates that are crucial for catalysis. The collection of modified proteins accrued during this work facilitated the generation and thorough characterization of monoclonal antibodies against tryptophan C-mannosylation. These antibodies empowered proteomic analyses of the brain C-glycome by enriching for peptides possessing tryptophan C-mannosylation. This study revealed many new modification sites on proteins throughout the secretory pathway with both conventional and non-canonical consensus sequences.


Subject(s)
Mannose/chemistry , Protein Engineering/methods , Tryptophan/metabolism , Amino Acid Sequence/genetics , Antibodies/immunology , Glycosylation , Glycosyltransferases/metabolism , Mannose/metabolism , Peptides/metabolism , Protein Processing, Post-Translational/physiology , Protein Stability , Proteomics/methods , Saccharomyces cerevisiae/metabolism , Saccharomycetales/metabolism , Tryptophan/chemistry
5.
Blood Adv ; 4(7): 1270-1283, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32236527

ABSTRACT

In eukaryotic cells, messenger RNA (mRNA) molecules are exported from the nucleus to the cytoplasm, where they are translated. The highly conserved protein nuclear RNA export factor1 (Nxf1) is an important mediator of this process. Although studies in yeast and in human cell lines have shed light on the biochemical mechanisms of Nxf1 function, its contribution to mammalian physiology is less clear. Several groups have identified recurrent NXF1 mutations in chronic lymphocytic leukemia (CLL), placing it alongside several RNA-metabolism factors (including SF3B1, XPO, RPS15) whose dysregulation is thought to contribute to CLL pathogenesis. We report here an allelic series of germline point mutations in murine Nxf1. Mice heterozygous for these loss-of-function Nxf1 mutations exhibit thrombocytopenia and lymphopenia, together with milder hematological defects. This is primarily caused by cell-intrinsic defects in the survival of platelets and peripheral lymphocytes, which are sensitized to intrinsic apoptosis. In contrast, Nxf1 mutations have almost no effect on red blood cell homeostasis. Comparative transcriptome analysis of platelets, lymphocytes, and erythrocytes from Nxf1-mutant mice shows that, in response to impaired Nxf1 function, the cytoplasmic representation of transcripts encoding regulators of RNA metabolism is altered in a unique, lineage-specific way. Thus, blood cell lineages exhibit differential requirements for Nxf1-mediated global mRNA export.


Subject(s)
Lymphopenia , Thrombocytopenia , Animals , Germ Cells , Lymphopenia/genetics , Mice , Mutation , Nucleocytoplasmic Transport Proteins/genetics , RNA, Viral , RNA-Binding Proteins/genetics , Thrombocytopenia/genetics
6.
mBio ; 11(1)2020 01 28.
Article in English | MEDLINE | ID: mdl-31992625

ABSTRACT

Activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) plays a critical role in antiviral responses to many DNA viruses. Sensing of cytosolic DNA by cGAS results in synthesis of the endogenous second messenger cGAMP that activates stimulator of interferon genes (STING) in infected cells. Critically, cGAMP can also propagate antiviral responses to uninfected cells through intercellular transfer, although the modalities of this transfer between epithelial and immune cells remain poorly defined. We demonstrate here that cGAMP-producing epithelial cells can transactivate STING in cocultured macrophages through direct cGAMP transfer. cGAMP transfer was reliant upon connexin expression by epithelial cells and pharmacological inhibition of connexins blunted STING-dependent transactivation of the macrophage compartment. Macrophage transactivation by cGAMP contributed to a positive-feedback loop amplifying antiviral responses, significantly protecting uninfected epithelial cells against viral infection. Collectively, our findings constitute the first direct evidence of a connexin-dependent cGAMP transfer to macrophages by epithelial cells, to amplify antiviral responses.IMPORTANCE Recent studies suggest that extracellular cGAMP can be taken up by macrophages to engage STING through several mechanisms. Our work demonstrates that connexin-dependent communication between epithelial cells and macrophages plays a significant role in the amplification of antiviral responses mediated by cGAMP and suggests that pharmacological strategies aimed at modulating connexins may have therapeutic applications to control antiviral responses in humans.


Subject(s)
Connexins/metabolism , Host-Pathogen Interactions , Nucleotides, Cyclic/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Animals , Biomarkers , Cells, Cultured , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Mice
7.
Nat Chem Biol ; 15(11): 1057-1066, 2019 11.
Article in English | MEDLINE | ID: mdl-31591564

ABSTRACT

Activating the intrinsic apoptosis pathway with small molecules is now a clinically validated approach to cancer therapy. In contrast, blocking apoptosis to prevent the death of healthy cells in disease settings has not been achieved. Caspases have been favored, but they act too late in apoptosis to provide long-term protection. The critical step in committing a cell to death is activation of BAK or BAX, pro-death BCL-2 proteins mediating mitochondrial damage. Apoptosis cannot proceed in their absence. Here we show that WEHI-9625, a novel tricyclic sulfone small molecule, binds to VDAC2 and promotes its ability to inhibit apoptosis driven by mouse BAK. In contrast to caspase inhibitors, WEHI-9625 blocks apoptosis before mitochondrial damage, preserving cellular function and long-term clonogenic potential. Our findings expand on the key role of VDAC2 in regulating apoptosis and demonstrate that blocking apoptosis at an early stage is both advantageous and pharmacologically tractable.


Subject(s)
Apoptosis/physiology , Small Molecule Libraries/metabolism , Voltage-Dependent Anion Channel 2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , Animals , Mice , Protein Binding , Voltage-Dependent Anion Channel 2/metabolism
8.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30760556

ABSTRACT

During platelet biogenesis, microtubules (MTs) are arranged into submembranous structures (the marginal band) that encircle the cell in a single plane. This unique MT array has no equivalent in any other mammalian cell, and the mechanisms responsible for this particular mode of assembly are not fully understood. One possibility is that platelet MTs are composed of a particular set of tubulin isotypes that carry specific posttranslational modifications. Although ß1-tubulin is known to be essential, no equivalent roles of α-tubulin isotypes in platelet formation or function have so far been reported. Here, we identify α4A-tubulin as a predominant α-tubulin isotype in platelets. Similar to ß1-tubulin, α4A-tubulin expression is up-regulated during the late stages of megakaryocyte differentiation. Missense mutations in the α4A-tubulin gene cause macrothrombocytopenia in mice and humans. Defects in α4A-tubulin lead to changes in tubulin tyrosination status of the platelet tubulin pool. Ultrastructural defects include reduced numbers and misarranged MT coils in the platelet marginal band. We further observed defects in megakaryocyte maturation and proplatelet formation in Tuba4a-mutant mice. We have, thus, discovered an α-tubulin isotype with specific and essential roles in platelet biogenesis.


Subject(s)
Blood Platelets/physiology , Thrombocytopenia/genetics , Thrombopoiesis/physiology , Tubulin/genetics , Tubulin/metabolism , Alkylating Agents/administration & dosage , Alkylating Agents/pharmacology , Animals , Antigens, CD34/metabolism , Cells, Cultured , Ethylnitrosourea/administration & dosage , Ethylnitrosourea/pharmacology , Humans , Male , Megakaryocytes/metabolism , Mice , Mice, Inbred BALB C , Microtubules/metabolism , Mutation, Missense , Platelet Count , Tissue Donors
9.
Nat Commun ; 9(1): 4976, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478310

ABSTRACT

Intrinsic apoptosis is critical to prevent tumor formation and is engaged by many anti-cancer agents to eliminate tumor cells. BAX and BAK, the two essential mediators of apoptosis, are thought to be regulated through similar mechanisms and act redundantly to drive apoptotic cell death. From an unbiased genome-wide CRISPR/Cas9 screen, we identified VDAC2 (voltage-dependent anion channel 2) as important for BAX, but not BAK, to function. Genetic deletion of VDAC2 abrogated the association of BAX and BAK with mitochondrial complexes containing VDAC1, VDAC2, and VDAC3, but only inhibited BAX apoptotic function. Deleting VDAC2 phenocopied the loss of BAX in impairing both the killing of tumor cells by anti-cancer agents and the ability to suppress tumor formation. Together, our studies show that efficient BAX-mediated apoptosis depends on VDAC2, and reveal a striking difference in how BAX and BAK are functionally impacted by their interactions with VDAC2.


Subject(s)
Apoptosis , Carcinogenesis/metabolism , Carcinogenesis/pathology , Voltage-Dependent Anion Channel 2/metabolism , bcl-2-Associated X Protein/metabolism , Animals , CRISPR-Cas Systems/genetics , Embryonic Development , HCT116 Cells , HeLa Cells , Humans , Mice, Inbred C57BL , Mitochondria/metabolism , Promoter Regions, Genetic/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
10.
Science ; 359(6378)2018 02 23.
Article in English | MEDLINE | ID: mdl-29472455

ABSTRACT

Mitochondrial apoptosis is mediated by BAK and BAX, two proteins that induce mitochondrial outer membrane permeabilization, leading to cytochrome c release and activation of apoptotic caspases. In the absence of active caspases, mitochondrial DNA (mtDNA) triggers the innate immune cGAS/STING pathway, causing dying cells to secrete type I interferon. How cGAS gains access to mtDNA remains unclear. We used live-cell lattice light-sheet microscopy to examine the mitochondrial network in mouse embryonic fibroblasts. We found that after BAK/BAX activation and cytochrome c loss, the mitochondrial network broke down and large BAK/BAX pores appeared in the outer membrane. These BAK/BAX macropores allowed the inner mitochondrial membrane to herniate into the cytosol, carrying with it mitochondrial matrix components, including the mitochondrial genome. Apoptotic caspases did not prevent herniation but dismantled the dying cell to suppress mtDNA-induced innate immune signaling.


Subject(s)
Apoptosis , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Cytochromes c/metabolism , DNA, Mitochondrial/metabolism , Fibroblasts , Gene Knockout Techniques , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mitochondrial Membranes/chemistry , Protein Multimerization , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
11.
Blood ; 131(6): 605-610, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29259001

ABSTRACT

Despite their profoundly different cellular composition, size, and function, megakaryocytes and platelets both depend on restraint of the intrinsic (or "mitochondrial") apoptosis pathway by BCL-2 family prosurvival proteins for their development and viability. Activation of the pathway contributes to the clearance of megakaryocytes following platelet shedding and constrains platelet lifespan in the circulation. Important questions remain as to how apoptosis is initiated in these cells at steady state and in response to pathophysiological insults.


Subject(s)
Apoptosis/physiology , Blood Platelets/physiology , Cell Lineage/physiology , Megakaryocytes/physiology , Animals , Cell Death , Cell Differentiation , Humans , Phagocytosis/physiology
12.
J Clin Invest ; 127(3): 814-829, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28134622

ABSTRACT

Platelets are anuclear cells that are essential for blood clotting. They are produced by large polyploid precursor cells called megakaryocytes. Previous genome-wide association studies in nearly 70,000 individuals indicated that single nucleotide variants (SNVs) in the gene encoding the actin cytoskeletal regulator tropomyosin 4 (TPM4) exert an effect on the count and volume of platelets. Platelet number and volume are independent risk factors for heart attack and stroke. Here, we have identified 2 unrelated families in the BRIDGE Bleeding and Platelet Disorders (BPD) collection who carry a TPM4 variant that causes truncation of the TPM4 protein and segregates with macrothrombocytopenia, a disorder characterized by low platelet count. N-Ethyl-N-nitrosourea-induced (ENU-induced) missense mutations in Tpm4 or targeted inactivation of the Tpm4 locus led to gene dosage-dependent macrothrombocytopenia in mice. All other blood cell counts in Tpm4-deficient mice were normal. Insufficient TPM4 expression in human and mouse megakaryocytes resulted in a defect in the terminal stages of platelet production and had a mild effect on platelet function. Together, our findings demonstrate a nonredundant role for TPM4 in platelet biogenesis in humans and mice and reveal that truncating variants in TPM4 cause a previously undescribed dominant Mendelian platelet disorder.


Subject(s)
Blood Platelets/metabolism , Genes, Dominant , Genetic Diseases, Inborn , Mutation, Missense , Thrombocytopenia , Tropomyosin , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genome-Wide Association Study , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Tropomyosin/genetics , Tropomyosin/metabolism
13.
Genes Dev ; 30(10): 1240-50, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198225

ABSTRACT

Due to the myriad interactions between prosurvival and proapoptotic members of the Bcl-2 family of proteins, establishing the mechanisms that regulate the intrinsic apoptotic pathway has proven challenging. Mechanistic insights have primarily been gleaned from in vitro studies because genetic approaches in mammals that produce unambiguous data are difficult to design. Here we describe a mutation in mouse and human Bak that specifically disrupts its interaction with the prosurvival protein Bcl-xL Substitution of Glu75 in mBak (hBAK Q77) for leucine does not affect the three-dimensional structure of Bak or killing activity but reduces its affinity for Bcl-xL via loss of a single hydrogen bond. Using this mutant, we investigated the requirement for physical restraint of Bak by Bcl-xL in apoptotic regulation. In vitro, Bak(Q75L) cells were significantly more sensitive to various apoptotic stimuli. In vivo, loss of Bcl-xL binding to Bak led to significant defects in T-cell and blood platelet survival. Thus, we provide the first definitive in vivo evidence that prosurvival proteins maintain cellular viability by interacting with and inhibiting Bak.


Subject(s)
Apoptosis/genetics , Blood Platelets/cytology , T-Lymphocytes/cytology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Line , Cell Survival/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Protein Binding , Protein Conformation , Protein Domains/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics
14.
Br J Haematol ; 174(6): 962-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27221652

ABSTRACT

Apoptosis is required to maintain tissue homeostasis in multicellular organisms. Platelets, the anucleate cells that are essential for blood clotting, are a prime example. Their brief life span in the circulation is regulated by the intrinsic apoptosis pathway. Pro-survival BCL-XL (also termed BCL2L1) is essential for platelet viability. It functions to restrain the pro-apoptotic BCL-2 family members BAK (also termed BAK1) and BAX, the essential mediators of intrinsic apoptosis. Genetic deletion or pharmacological inhibition of BCL-XL results in thrombocytopenia. Conversely, deletion of BAK in platelets doubles their circulating life span. However, what triggers platelet apoptosis in vivo remains unclear. The pro-apoptotic BH3-only proteins are essential for initiating apoptosis in nucleated cells, and there is some evidence to suggest they also play a role in platelet biology. We investigated whether PUMA (also termed BBC3), a potent BH3-only protein that can inhibit all pro-survival BCL-2 family members as well as directly activate BAX, regulates the death of platelets. Surprisingly, loss of PUMA had no impact on the loss of platelets caused by loss of BCL-XL. It therefore remains to be established whether other BH3-only proteins play a critical role in induction of apoptosis in platelets or whether their death is controlled solely by the interactions between BCL-XL with BAK and BAX.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Gene Deletion , Genetic Association Studies , Genetic Predisposition to Disease , Proto-Oncogene Proteins/deficiency , Thrombocytopenia/genetics , bcl-2-Associated X Protein/deficiency , Animals , Apoptosis Regulatory Proteins/genetics , Blood Platelets/metabolism , Disease Models, Animal , Gene Expression , Genotype , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Thrombocytopenia/blood , bcl-2-Associated X Protein/genetics
15.
eNeuro ; 3(1)2016.
Article in English | MEDLINE | ID: mdl-27022620

ABSTRACT

Type-1 interferons (IFNs) are pleiotropic cytokines that signal through the type-1 IFN receptor (IFNAR1). Recent literature has implicated the type-1 IFNs in disorders of the CNS. In this study, we have investigated the role of type-1 IFNs in neuroinflammation following traumatic brain injury (TBI). Using a controlled cortical impact model, TBI was induced in 8- to 10-week-old male C57BL/6J WT and IFNAR1(-/-) mice and brains were excised to study infarct volume, inflammatory mediator release via quantitative PCR analysis and immune cell profile via immunohistochemistry. IFNAR1(-/-) mice displayed smaller infarcts compared with WT mice after TBI. IFNAR1(-/-) mice exhibited an altered anti-inflammatory environment compared with WT mice, with significantly reduced levels of the proinflammatory mediators TNFα, IL-1ß and IL-6, an up-regulation of the anti-inflammatory mediator IL-10 and an increased activation of resident and peripheral immune cells after TBI. WT mice injected intravenously with an anti-IFNAR1 blocking monoclonal antibody (MAR1) 1 h before, 30 min after or 30 min and 2 d after TBI displayed significantly improved histological and behavioral outcome. Bone marrow chimeras demonstrated that the hematopoietic cells are a peripheral source of type-1 IFNs that drives neuroinflammation and a worsened TBI outcome. Type-1 IFN mRNA levels were confirmed to be significantly altered in human postmortem TBI brains. Together, these data demonstrate that type-1 IFN signaling is a critical pathway in the progression of neuroinflammation and presents a viable therapeutic target for the treatment of TBI.


Subject(s)
Brain Injuries/metabolism , Encephalitis/metabolism , Hematopoietic Stem Cells/metabolism , Interferon Type I/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Brain Injuries/complications , Brain Injuries/pathology , Encephalitis/etiology , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction
16.
Blood ; 127(14): 1743-51, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26773046

ABSTRACT

Human platelets contain microRNAs (miRNAs) and miRNA processing machinery, but their contribution to platelet function remains incompletely understood. Here, we show that murine megakaryocyte (MK)-specific knockdown of Dicer1, the ribonuclease that cleaves miRNA precursors into mature miRNAs, reduces the level of the majority of miRNAs in platelets. This leads to altered platelet messenger RNA (mRNA) expression profiles and mild thrombocytopenia. Fibrinogen receptor subunits Itga2b (αIIb) and Itgb3 (ß3) mRNAs were among the differentially expressed transcripts that are increased in platelets lacking Dicer1. Argonaute 2 (Ago2), a member of the miRNA silencing complex, co-immunoprecipitated with αIIband ß3mRNAs in wild-type platelets. Furthermore, co-immunoprecipitation experiments suggested reduced αIIb/ß3/Ago2 complexes in miRNA-deficient platelets. These results suggested that miRNAs regulate both integrin subunits. Subsequent 3' untranslated region luciferase reporter assays confirmed that the translation of both αIIband ß3mRNAs can be regulated by miRNAs miR-326, miR-128, miR-331, and miR-500. Consistent with these molecular changes, the deletion ofDicer1resulted in increased surface expression of integrins αIIband ß3, and enhanced platelet binding to fibrinogen in vivo and in vitro. Heightened platelet reactivity, shortened tail-bleeding time, and reduced survival following collagen/epinephrine-induced pulmonary embolism were also observed in Dicer1-deficient animals. CombinedPf4-cre-mediated deletion of Drosha and Dicer1 did not significantly exacerbate phenotypes observed in single Dicer1 knockout mice. In summary, these findings indicate that Dicer1-dependent generation of mature miRNAs in late-stage MKs and platelets modulates the expression of target mRNAs important for the hemostatic and thrombotic function of platelets.


Subject(s)
Blood Platelets/metabolism , DEAD-box RNA Helicases/metabolism , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Ribonuclease III/metabolism , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , DEAD-box RNA Helicases/genetics , Humans , Integrin alpha2/biosynthesis , Integrin alpha2/genetics , Integrin beta3/biosynthesis , Integrin beta3/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Pulmonary Embolism/chemically induced , Pulmonary Embolism/genetics , Pulmonary Embolism/metabolism , RNA, Messenger/genetics , Ribonuclease III/genetics
17.
Proc Natl Acad Sci U S A ; 111(35): 12835-40, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136120

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) have emerged as important cellular players in tissue repair and innate immunity. Whether these cells meaningfully regulate adaptive immune responses upon activation has yet to be explored. Here we show that upon IL-1ß stimulation, peripheral ILC3s become activated, secrete cytokines, up-regulate surface MHC class II molecules, and express costimulatory molecules. ILC3s can take up latex beads, process protein antigen, and consequently prime CD4(+) T-cell responses in vitro. The cognate interaction of ILC3s and CD4(+) T cells leads to T-cell proliferation both in vitro and in vivo, whereas its disruption impairs specific T-cell and T-dependent B-cell responses in vivo. In addition, the ILC3-CD4(+) T-cell interaction is bidirectional and leads to the activation of ILC3s. Taken together, our data reveal a novel activation-dependent function of peripheral ILC3s in eliciting cognate CD4(+) T-cell immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-1beta/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Signal Transduction/immunology , Animals , Antigen Presentation/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Histocompatibility Antigens Class II/immunology , Immunity, Cellular/immunology , Interleukin-1beta/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL
18.
J Immunol ; 189(8): 3894-904, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22972926

ABSTRACT

Thymic epithelial cells provide unique cues for the lifelong selection and differentiation of a repertoire of functionally diverse T cells. Rendered microRNA (miRNA) deficient, these stromal cells in the mouse lose their capacity to instruct the commitment of hematopoietic precursors to a T cell fate, to effect thymocyte positive selection, and to achieve promiscuous gene expression required for central tolerance induction. Over time, the microenvironment created by miRNA-deficient thymic epithelia assumes the cellular composition and structure of peripheral lymphoid tissue, where thympoiesis fails to be supported. These findings emphasize a global role for miRNA in the maintenance and function of the thymic epithelial cell scaffold and establish a novel mechanism how these cells control peripheral tissue Ag expression to prompt central immunological tolerance.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , MicroRNAs/physiology , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , DEAD-box RNA Helicases/deficiency , Epithelial Cells/pathology , Mice , Mice, Nude , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Mutation , Organ Culture Techniques , Ribonuclease III/deficiency , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/embryology
19.
PLoS One ; 7(2): e31939, 2012.
Article in English | MEDLINE | ID: mdl-22384106

ABSTRACT

Interleukin-7 (IL-7) is a major survival factor for mature T cells. Therefore, the degree of IL-7 availability determines the size of the peripheral T cell pool and regulates T cell homeostasis. Here we provide evidence that IL-7 also regulates the homeostasis of intestinal epithelial cells (IEC), colon function and the composition of the commensal microflora. In the colon of T cell-deficient, lymphopenic mice, IL-7-producing IEC accumulate. IEC hyperplasia can be blocked by IL-7-consuming T cells or the inactivation of the IL-7/IL-7R signaling pathway. However, the blockade of the IL-7/IL-7R signaling pathway renders T cell-deficient mice more sensitive to chemically-induced IEC damage and subsequent colitis. In summary, our data demonstrate that IL-7 promotes IEC hyperplasia under lymphopenic conditions. Under non-lymphopenic conditions, however, T cells consume IL-7 thereby limiting IEC expansion and survival. Hence, the degree of IL-7 availability regulates both, T cell and IEC homeostasis.


Subject(s)
Epithelial Cells/cytology , Interleukin-7/metabolism , Intestines/cytology , T-Lymphocytes/cytology , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Colitis/metabolism , Genes, RAG-1/genetics , Homeostasis , Hyperplasia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, Interleukin-7/metabolism , Signal Transduction
20.
J Immunol ; 185(6): 3514-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709954

ABSTRACT

Hematopoietic lymphoid tissue inducer (LTi) cells initiate lymph node (LN) and Peyer's patch (PP) development during fetal life by inducing the differentiation of mesenchymal organizer cells. The growth factor signals underlying LTi cell development and LN and PP organogenesis remain poorly understood. LTi cells express the Il7r and the receptor tyrosine kinase Kit, whereas organizer cells express their cognate ligands. To determine the relative significance of Il7 and Kit signaling in LTi cell homeostasis and PP and LN development, we have analyzed mice deficient for Kit (Kit(W/Wv)), Il7 (Il7(-/-)), or both (Il7(-/-) Kit(W/Wv)). Unlike Kit(W/Wv) and Il7(-/-) single mutants, Il7(-/-) Kit(W/Wv) mice were almost devoid of LTi cells in their mesenteric LN anlage. This LTi deficiency was associated with a block in mesenchymal LN organizer cell generation and the absence of almost all LNs. In contrast, intestinal LTi cell numbers, PP organizer cell generation, and PP development were strongly affected by impaired Kit signaling, but were independent of Il7. Hence, Kit and Il7 act synergistically in LN organogenesis, whereas Kit signaling, but not Il7, critically regulates PP organogenesis and LTi cell numbers in the intestine. Consistent with these differential growth factor requirements for PP and LN development, PP organizer cells expressed higher Kitl and lower Il7 levels than did LN organizer cells. Collectively, these results demonstrate that Kit and Il7 differentially control PP and LN organogenesis through the local growth factor-driven regulation of LTi cell numbers.


Subject(s)
Cell Differentiation/immunology , Interleukin-7/physiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Peyer's Patches/cytology , Peyer's Patches/immunology , Stem Cell Factor/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Interleukin-7/deficiency , Interleukin-7/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/immunology , Lymph Nodes/embryology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/genetics , Organogenesis/immunology , Peyer's Patches/embryology , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...