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










Publication year range
1.
Gut ; 73(1): 92-104, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37595983

ABSTRACT

OBJECTIVE: Wheat has become a main staple globally. We studied the effect of defined pro-inflammatory dietary proteins, wheat amylase trypsin inhibitors (ATI), activating intestinal myeloid cells via toll-like receptor 4, in experimental autoimmune encephalitis (EAE), a model of multiple sclerosis (MS). DESIGN: EAE was induced in C57BL/6J mice on standardised dietary regimes with defined content of gluten/ATI. Mice received a gluten and ATI-free diet with defined carbohydrate and protein (casein/zein) content, supplemented with: (a) 25% of gluten and 0.75% ATI; (b) 25% gluten and 0.19% ATI or (c) 1.5% purified ATI. The effect of dietary ATI on clinical EAE severity, on intestinal, mesenteric lymph node, splenic and central nervous system (CNS) subsets of myeloid cells and lymphocytes was analysed. Activation of peripheral blood mononuclear cells from patients with MS and healthy controls was compared. RESULTS: Dietary ATI dose-dependently caused significantly higher EAE clinical scores compared with mice on other dietary regimes, including on gluten alone. This was mediated by increased numbers and activation of pro-inflammatory intestinal, lymph node, splenic and CNS myeloid cells and of CNS-infiltrating encephalitogenic T-lymphocytes. Expectedly, ATI activated peripheral blood monocytes from both patients with MS and healthy controls. CONCLUSIONS: Dietary wheat ATI activate murine and human myeloid cells. The amount of ATI present in an average human wheat-based diet caused mild intestinal inflammation, which was propagated to extraintestinal sites, leading to exacerbation of CNS inflammation and worsening of clinical symptoms in EAE. These results support the importance of the gut-brain axis in inflammatory CNS disease.


Subject(s)
Multiple Sclerosis , Humans , Animals , Mice , Trypsin Inhibitors/pharmacology , Trypsin Inhibitors/chemistry , Triticum/chemistry , Amylases , Leukocytes, Mononuclear , Mice, Inbred C57BL , Inflammation , Central Nervous System , Glutens , Diet
2.
J Invest Dermatol ; 143(7): 1257-1267.e10, 2023 07.
Article in English | MEDLINE | ID: mdl-36736996

ABSTRACT

Keratinocytes (KCs) form the outer epithelial barrier of the body, protecting against invading pathogens. Mice lacking the IL-17RA or both IL-17A and IL-17F develop spontaneous Staphylococcusaureus skin infections. We found a marked expansion of T17 cells, comprised of RORγt-expressing γδ T cells and T helper 17 cells in the skin-draining lymph nodes of these mice. Contradictory to previous suggestions, this expansion was not a result of a direct negative feedback loop because we found no expansion of T17 cells in mice lacking IL-17 signaling specifically in T cells. Instead, we found that the T17 expansion depended on the microbiota and was observed only when KCs were deficient for IL-17RA signaling. Indeed, mice that lack IL-17RA only in KCs showed an increased susceptibility to experimental epicutaneous infection with S. aureus together with an accumulation of IL-17A-producing γδ T cells. We conclude that deficiency of IL-17RA on KCs leads to microbiota dysbiosis in the skin, which triggers the expansion of IL-17A-producing T cells. Our data show that KCs are the primary target cells of IL-17A and IL-17F, coordinating the defense against microbial invaders in the skin.


Subject(s)
Interleukin-17 , Staphylococcus aureus , Mice , Animals , Mice, Knockout , Skin , Keratinocytes , Mice, Inbred C57BL
3.
Toxicol Sci ; 189(2): 237-249, 2022 09 24.
Article in English | MEDLINE | ID: mdl-35737426

ABSTRACT

SLN360 is a liver-targeted N-acetyl galactosamine (GalNAc)-conjugated small interfering RNA (siRNA) with a promising profile for addressing lipoprotein (a)-related cardiovascular risk. Here, we describe the findings from key preclinical safety studies. In vitro, SLN360 specifically reduced LPA expression in primary human hepatocytes with no relevant off-target effects. In rats, 10 mg/kg subcutaneous SLN360 was distributed specifically to the liver and kidney (peak 126 or 246 mg/g tissue at 6 h, respectively), with <1% of peak liver levels observed in all other tested organs. In vitro, no genotoxicity and no effect on human Ether-a-go-go Related Gene currents or proinflammatory cytokine production was observed, whereas in vivo, no SLN360-specific antibodies were detected in rabbit serum. In rat and nonhuman primate 29-day toxicology studies, SLN360 was well tolerated at all doses. In both species, known GalNAc-conjugated siRNA-induced microscopic changes were observed in the kidney and liver, with small increases in alanine aminotransferase and alkaline phosphatase observed in the high dose rats. Findings were in line with previously described siRNA-GalNAc platform-related effects and all observations were reversible and considered nonadverse. In cynomolgus monkeys, liver LPA messenger RNA and serum lipoprotein (a) were significantly reduced at day 30 and after an 8-week recovery period. No dose-related changes in safety assessment endpoints were noted. No SLN360-induced cytokine production, complement activation, or micronucleus formation was observed in vivo. The toxicological profile of SLN360 presented here is restricted to known GalNAc siRNA effects and no other toxicity associated with SLN360 has been noted. The preclinical profile of SLN360 confirmed suitability for entry into clinical studies.


Subject(s)
Acetylgalactosamine , Cardiovascular Diseases , Acetylgalactosamine/metabolism , Acetylgalactosamine/toxicity , Alanine Transaminase , Alkaline Phosphatase , Animals , Cytokines , Ethers , Humans , Lipoprotein(a) , Macaca fascicularis , RNA, Messenger , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rabbits , Rats
4.
Atherosclerosis ; 349: 240-247, 2022 05.
Article in English | MEDLINE | ID: mdl-35400495

ABSTRACT

BACKGROUND AND AIMS: The LPA gene encodes apolipoprotein (a), a key component of Lp(a), a potent risk factor for cardiovascular disease with no specific pharmacotherapy. Here we describe the pharmacological data for SLN360, a GalNAc-conjugated siRNA targeting LPA, designed to address this unmet medical need. METHODS: SLN360 was tested in vitro for LPA knockdown in primary hepatocytes. Healthy cynomolgus monkeys received single or multiple subcutaneous doses of the SLN360 sequence ranging from 0.1 to 9.0 mg/kg to determine the pharmacokinetic and pharmacodynamic effects. Liver mRNA and serum biomarker analyses were performed. RESULTS: In vitro, the SLN360 sequence potently reduces LPA mRNA in primary cynomolgus and human hepatocytes, while no effect was observed on the expression of APOB or PLG. In vivo, SLN360 exposure peaks 2 h after subcutaneous injection with near full elimination by 24 h. Specific LPA mRNA reduction (up to 91% 2 weeks after dosing) was observed with only the 3 mg/kg group showing appreciable return to baseline (40%). No consistent dose- or time-dependent effect on the expression of APOB, PLG or a panel of sensitive markers of liver lipid accumulation was observed. Potent (up to 95%) and long lasting (≥9 weeks) serum Lp(a) reduction was observed, peaking in all active groups at day 21. The minimally effective dose was determined to be 0.3 mg/kg with an ED50 of 0.6 mg/kg. CONCLUSIONS: SLN360 induces a sustained reduction in serum Lp(a) levels in cynomolgus monkeys following subcutaneous dosing. SLN360 has potential to address the unmet need of Lp(a) reduction in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Hyperlipidemias , Apolipoproteins A , Apolipoproteins B , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Humans , Lipoprotein(a) , RNA, Messenger , RNA, Small Interfering/genetics
5.
Mol Ther Nucleic Acids ; 27: 1116-1126, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35251767

ABSTRACT

Synthetic siRNA guide strands are typically designed with perfect complementarity to the passenger strand and the target mRNA. We examined whether siRNAs with intentional guide-strand bulges are functional in vitro and in vivo. Importantly, this was done by systematic shortening of the passenger strand, evaluating identical 19-mer guide-strand sequences but forcing them into conformations with 1- to 4-nt bulges after annealing. We demonstrate that guide-strand bulges can be well tolerated at several positions of unmodified and modified siRNAs. Beyond that, we show that GalNAc-conjugated siRNAs with bulges at certain positions of the guide strand repress transthyretin in murine primary hepatocytes and in vivo in mice. In vivo, a GalNAc-conjugated siRNA with a 1-nt bulge at position 14 of the guide strand was as active as the perfectly complementary siRNA. Finally, in a luciferase reporter system, mRNA target sequences were systematically shortened so that RNA-induced silencing complex activity could only occur with a guide-strand bulge. Here, luciferase reporters were repressed when 1- and 2-nt deletions of the reporter were applied to the edges of the sequence. We conclude that some guide-strand bulges versus target transcript can result in target repression and therefore should be evaluated as off-target risks.

6.
Sci Immunol ; 6(56)2021 02 05.
Article in English | MEDLINE | ID: mdl-33547052

ABSTRACT

Interleukin-17A- (IL-17A) and IL-17F-producing CD4+ T helper cells (TH17 cells) are implicated in the development of chronic inflammatory diseases, such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). TH17 cells also orchestrate leukocyte invasion of the central nervous system (CNS) and subsequent tissue damage. However, the role of IL-17A and IL-17F as effector cytokines is still confused with the encephalitogenic function of the cells that produce these cytokines, namely, TH17 cells, fueling a long-standing debate in the neuroimmunology field. Here, we demonstrated that mice deficient for IL-17A/F lose their susceptibility to EAE, which correlated with an altered composition of their gut microbiota. However, loss of IL-17A/F in TH cells did not diminish their encephalitogenic capacity. Reconstitution of a wild-type-like intestinal microbiota or reintroduction of IL-17A specifically into the gut epithelium of IL-17A/F-deficient mice reestablished their susceptibility to EAE. Thus, our data demonstrated that IL-17A and IL-17F are not encephalitogenic mediators but rather modulators of intestinal homeostasis that indirectly alter CNS-directed autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome/immunology , Interleukin-17/metabolism , Multiple Sclerosis/immunology , Adoptive Transfer , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/microbiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Fecal Microbiota Transplantation , Female , Humans , Interleukin-17/genetics , Male , Mice , Mice, Knockout , Multiple Sclerosis/pathology , Th17 Cells/immunology , Th17 Cells/transplantation
7.
Acta Neuropathol ; 140(4): 549-567, 2020 10.
Article in English | MEDLINE | ID: mdl-32651669

ABSTRACT

The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.


Subject(s)
Blood-Brain Barrier/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/enzymology , Heme Oxygenase-1/metabolism , Inflammation/immunology , Interleukin-1/immunology , Animals , Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
8.
J Mol Med (Berl) ; 96(8): 819-829, 2018 08.
Article in English | MEDLINE | ID: mdl-29959474

ABSTRACT

IL-17A and IL-17F share the highest sequence homology of the IL-17 family and signal via the same IL-17RA/RC receptor heterodimer. To better explore the expression of these two cytokines, we used a double reporter mouse strain (IL-17DR mice), where IL-17A expressing cells are marked by enhanced green fluorescent protein (eGFP) while red fluorescence protein (RFP) reports the expression of IL-17F. In steady state, we found that Th17 and γδ T cells only expressed IL-17A, while IL-17F expression was restricted to CD8 T cells (Tc17) and innate lymphoid cells (ILC type 3) of the gut. In experimental autoimmune encephalomyelitis, the vast majority of CNS-infiltrating Th17 cells expressed IL-17A but not IL-17F. In contrast, anti-CD3-induced, TGF-ß-driven Th17 cells in the gut expressed both of these IL-17 cytokines. In line with this, in vitro differentiation of Th17 cells in the presence of IL-1ß led primarily to IL-17A expressing T cells, while TGF-ß induced IL-17F co-expressing Th17 cells. Our results suggest that expression of IL-17F is associated with non-pathogenic T cells, pointing to a differential function of IL-17A versus IL-17F. KEY MESSAGES: Naïve mice: CD4+ T cells and γδ T cells express IL-17A, and Tc17 cells express IL-17F. Gut ILC3 show differential expression of IL17A and F. Th17 differentiation with TGF-ß1 induces IL-17A and F, whereas IL-1ß induced cells expressing IL-17A. Th17 cells in EAE in CNS express IL-17A only. Gut Th17 cells induced by anti-CD3 express IL-17A and F together as skin γδ T cells of IMQ-treated mice.


Subject(s)
Gene Expression , Interleukin-17/genetics , Th17 Cells/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental , Immunophenotyping , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology , Th17 Cells/immunology
9.
EMBO J ; 36(14): 2088-2106, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28645918

ABSTRACT

Argonaute proteins associate with microRNAs and are key components of gene silencing pathways. With such a pivotal role, these proteins represent ideal targets for regulatory post-translational modifications. Using quantitative mass spectrometry, we find that a C-terminal serine/threonine cluster is phosphorylated at five different residues in human and Caenorhabditis elegans In human, hyper-phosphorylation does not affect microRNA binding, localization, or cleavage activity of Ago2. However, mRNA binding is strongly affected. Strikingly, on Ago2 mutants that cannot bind microRNAs or mRNAs, the cluster remains unphosphorylated indicating a role at late stages of gene silencing. In C. elegans, the phosphorylation of the conserved cluster of ALG-1 is essential for microRNA function in vivo Furthermore, a single point mutation within the cluster is sufficient to phenocopy the loss of its complete phosphorylation. Interestingly, this mutant retains its capacity to produce and bind microRNAs and represses expression when artificially tethered to an mRNA Altogether, our data suggest that the phosphorylation state of the serine/threonine cluster is important for Argonaute-mRNA interactions.


Subject(s)
Argonaute Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Gene Silencing , MicroRNAs/metabolism , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Animals , Argonaute Proteins/genetics , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Humans , Phosphorylation , Protein Binding
10.
Methods Mol Biol ; 1580: 107-116, 2017.
Article in English | MEDLINE | ID: mdl-28439830

ABSTRACT

Argonaute (Ago) proteins bind small RNAs such as microRNAs (miRNAs) or short interfering RNAs (siRNAs), which guide them to distinct mRNAs for post-transcriptional gene silencing. Mammalian miRNA-guided gene silencing pathways mainly lead to translational repression and mRNA destabilization. To facilitate these processes, Ago proteins bind members of the GW protein family, which form central interaction platforms for the recruitment of downstream effector proteins. GW proteins use tryptophane residues (W) to bind to the surface of Ago proteins. This high affinity interaction is retained when a short, GST-fused GW peptide is used in biochemical pull-down experiments-an approach referred to as "Ago Affinity Purification by Peptides" (Ago-APP). Since the binding interface is conserved among different paralogues and different species, Ago-APP represents a universal tool to purify Ago proteins and associated small RNAs using samples from species with conserved miRNA pathways.


Subject(s)
Argonaute Proteins/isolation & purification , Peptides/metabolism , Animals , Argonaute Proteins/metabolism , Blotting, Northern/methods , Cell Culture Techniques/methods , Humans , Peptides/isolation & purification , Protein Binding , Protein Interaction Mapping/methods , RNA, Small Interfering/isolation & purification , RNA, Small Interfering/metabolism
11.
Plant Mol Biol ; 93(3): 283-298, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28004241

ABSTRACT

KEY MESSAGE: We identify proteins that associate with the THO core complex, and show that the TEX1 and MOS11 components functionally interact, affecting mRNA export and splicing as well as plant development. TREX (TRanscription-EXport) is a multiprotein complex that plays a central role in the coordination of synthesis, processing and nuclear export of mRNAs. Using targeted proteomics, we identified proteins that associate with the THO core complex of Arabidopsis TREX. In addition to the RNA helicase UAP56 and the mRNA export factors ALY2-4 and MOS11 we detected interactions with the mRNA export complex TREX-2 and multiple spliceosomal components. Plants defective in the THO component TEX1 or in the mRNA export factor MOS11 (orthologue of human CIP29) are mildly affected. However, tex1 mos11 double-mutant plants show marked defects in vegetative and reproductive development. In tex1 plants, the levels of tasiRNAs are reduced, while miR173 levels are decreased in mos11 mutants. In nuclei of mos11 cells increased mRNA accumulation was observed, while no mRNA export defect was detected with tex1 cells. Nevertheless, in tex1 mos11 double-mutants, the mRNA export defect was clearly enhanced relative to mos11. The subnuclear distribution of TEX1 substantially overlaps with that of splicing-related SR proteins and in tex1 plants the ratio of certain alternative splicing events is altered. Our results demonstrate that Arabidopsis TEX1 and MOS11 are involved in distinct steps of the biogenesis of mRNAs and small RNAs, and that they interact regarding some aspects, but act independently in others.


Subject(s)
Alternative Splicing/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , RNA Transport , RNA-Binding Proteins/metabolism , Arabidopsis/ultrastructure , Arabidopsis Proteins/genetics , Arabidopsis Proteins/isolation & purification , Inflorescence/metabolism , Inflorescence/ultrastructure , Models, Biological , Phenotype , Protein Binding , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Plant/metabolism , RNA-Binding Proteins/genetics
12.
EMBO J ; 36(1): 102-115, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27827809

ABSTRACT

Interleukin-1 (IL-1) is implicated in numerous pathologies, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). However, the exact mechanism by which IL-1 is involved in the generation of pathogenic T cells and in disease development remains largely unknown. We found that following EAE induction, pertussis toxin administration leads to IL-1 receptor type 1 (IL-1R1)-dependent IL-1ß expression by myeloid cells in the draining lymph nodes. This myeloid-derived IL-1ß did not vitally contribute to the generation and plasticity of Th17 cells, but rather promoted the expansion of a GM-CSF+ Th17 cell subset, thereby enhancing its encephalitogenic potential. Lack of expansion of GM-CSF-producing Th17 cells led to ameliorated disease in mice deficient for IL-1R1 specifically in T cells. Importantly, pathogenicity of IL-1R1-deficient T cells was fully restored by IL-23 polarization and expansion in vitro Therefore, our data demonstrate that IL-1 functions as a mitogenic mediator of encephalitogenic Th17 cells rather than qualitative inducer of their generation.


Subject(s)
Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Interleukin-1/metabolism , Th17 Cells/chemistry , Th17 Cells/physiology , Animals , Mice , Pertussis Toxin/administration & dosage , Pertussis Toxin/toxicity
13.
PLoS One ; 11(8): e0161165, 2016.
Article in English | MEDLINE | ID: mdl-27518285

ABSTRACT

The members of the Argonaute (AGO) protein family are key players in miRNA-guided gene silencing. They enable the interaction between small RNAs and their respective target mRNA(s) and support the catalytic destruction of the gene transcript or recruit additional proteins for downstream gene silencing. The human AGO family consists of four AGO proteins (AGO1-AGO4), but only AGO2 harbors nuclease activity. In this study, we characterized the expression of the four AGO proteins in cancer cell lines and normal tissues with a new mass spectrometry approach called AGO-APP (AGO Affinity Purification by Peptides). In all analyzed normal tissues, AGO1 and AGO2 were most prominent, but marked tissue-specific differences were identified. Furthermore, considerable changes during development were observed by comparing fetal and adult tissues. We also identified decreased overall AGO expression in melanoma derived cell lines compared to other tumor cell lines and normal tissues, with the largest differences in AGO2 expression. The experiments described in this study suggest that reduced amounts of AGO proteins, as key players in miRNA processing, have impact on several cellular processes. Deregulated miRNA expression has been attributed to chromosomal aberrations, promoter regulation and it is known to have a major impact on tumor development and progression. Our findings will further increase our basic understanding of the molecular basis of miRNA processing and its relevance for disease.


Subject(s)
Argonaute Proteins/metabolism , Fetus/metabolism , Gene Expression Regulation, Developmental , Melanoma/metabolism , Skin Neoplasms/metabolism , Skin/metabolism , Argonaute Proteins/genetics , Blotting, Western , Cells, Cultured , Humans , Melanoma/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Plant Cell ; 28(7): 1563-80, 2016 07.
Article in English | MEDLINE | ID: mdl-27354557

ABSTRACT

ARGONAUTE1 (AGO1) mediates posttranscriptional silencing by microRNAs (miRNAs) and short interfering RNAS (siRNAs). AGO1-catalyzed RNA cleavage (slicing) represses miRNA targets, but current models also highlight the roles of slicing in formation of siRNAs and siRNA-AGO1 complexes. miRNA-guided slicing is required for biogenesis of phased, trans-acting siRNAs (tasiRNAs), whose cleaved precursor fragments are converted to double-stranded RNA by RNA-dependent RNA polymerase 6 (RDR6). In addition, unwinding of duplex siRNA bound to AGO1 requires passenger strand cleavage in vitro. In this study, we analyze how mutation of four metal ion-coordinating residues of Arabidopsis thaliana AGO1 affects slicer activity in vitro and siRNA function in vivo. We show that while all four residues are required for slicer activity, they do not contribute equally to catalysis. Moreover, passenger strand cleavage is required for assembly of active AGO1-siRNA complexes in vivo, and many AGO1-bound siRNAs are trimmed in the absence of slicer activity. Remarkably, seedlings defective in AGO1 slicer activity produce abundant siRNAs from tasiRNA loci in vivo. These siRNAs depend on RDR6 and SUPPRESSOR OF GENE SILENCING3, but unlike wild-type tasiRNAs, they are unphased. These results demonstrate that slicing is solely required for phase definition of tasiRNAs, and they strongly support recruitment of RDR6 by AGO1 rather than by cleavage fragments.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Argonaute Proteins/metabolism , MicroRNAs/genetics , RNA, Plant/genetics , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Argonaute Proteins/genetics , RNA, Small Interfering/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
15.
Proc Natl Acad Sci U S A ; 112(38): 11841-5, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26351695

ABSTRACT

During microRNA (miRNA)-guided gene silencing, Argonaute (Ago) proteins interact with a member of the TNRC6/GW protein family. Here we used a short GW protein-derived peptide fused to GST and demonstrate that it binds to Ago proteins with high affinity. This allows for the simultaneous isolation of all Ago protein complexes expressed in diverse species to identify associated proteins, small RNAs, or target mRNAs. We refer to our method as "Ago protein Affinity Purification by Peptides" (Ago-APP). Furthermore, expression of this peptide competes for endogenous TNRC6 proteins, leading to global inhibition of miRNA function in mammalian cells.


Subject(s)
Argonaute Proteins/isolation & purification , Chromatography, Affinity/methods , Multiprotein Complexes/isolation & purification , Peptides/isolation & purification , Amino Acid Sequence , Animals , Cell Extracts , Chemical Precipitation , Drosophila melanogaster , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , MicroRNAs/metabolism , Molecular Sequence Data , Peptides/chemistry
16.
Acta Neuropathol ; 129(5): 625-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25716179

ABSTRACT

Cytokines of the IL-17 family are uniquely placed on the border between immune cells and tissue. Although IL-17 was originally found to induce the activation and mobilization of neutrophils to sites of inflammation, its tissue-specific function is not yet fully understood. The best-studied IL-17 family members, IL-17A and IL-17F, are both typically produced by immune cells such as Th17, γδ T cells and innate lymphoid cells group 3. However, the cells that respond to these cytokines are mostly found in inflamed tissue. As seen in psoriatic skin lesions or in joints of rheumatoid arthritis patients, high levels of IL-17 have been detected in the central nervous system (CNS) during inflammatory responses. Here, we provide a general review of the molecular function of IL-17 and its role in the CNS in particular. Of the different inflammatory conditions of the CNS, we found multiple sclerosis (MS) to be the one most associated with the presence of Th17 cells and IL-17. In particular, many studies using the murine model for MS, experimental autoimmune encephalomyelitis, found a clear association of Th17 and IL-17 with disease severity and progression. We summarize the recent advances made in correlating the presence of IL-17 with impaired blood-brain barrier integrity as well as the activation of astrocytes and microglia and the consequences for disease progression. There is also evidence that IL-17 plays a pathogenic role in the post-ischemic phase of stroke as well as its experimental model. We review the limited but promising data on the sources of post-stroke IL-17 production and its effects on CNS-resident target cells. In addition to MS and stroke, there is also evidence linking high levels of IL-17 to depression, as a frequent comorbidity of several inflammatory diseases, as well as to different types of infections of the CNS. The evidence we supply here suggests that inhibiting the function of the IL-17 cytokine family could have a beneficial effect on pathogenic conditions in the CNS.


Subject(s)
Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Inflammation/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Animals , Astrocytes/cytology , Astrocytes/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Depression/immunology , Depression/pathology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Inflammation/pathology , Interleukin-17/classification , Microglia/cytology , Microglia/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Risk Factors , Stroke/immunology , Stroke/pathology
17.
RNA ; 20(10): 1532-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25114291

ABSTRACT

Argonaute proteins bind small RNAs and mediate cleavage of complementary target RNAs. The human Argonaute protein Ago4 is catalytically inactive, although it is highly similar to catalytic Ago2. Here, we have generated Ago2-Ago4 chimeras and analyzed their cleavage activity in vitro. We identify several specific features that inactivate Ago4: the catalytic center, short sequence elements in the N-terminal domain, and an Ago4-specific insertion in the catalytic domain. In addition, we show that Ago2-mediated cleavage of the noncanonical miR-451 precursor can be carried out by any catalytic human Ago protein. Finally, phylogenetic analyses establish evolutionary distances between the Ago proteins. Interestingly, these distances do not fully correlate with the structural changes inactivating them, suggesting functional adaptations of individual human Ago proteins.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , RNA Cleavage/genetics , Amino Acid Sequence , Argonaute Proteins/metabolism , Blotting, Northern , Catalysis , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Humans , MicroRNAs/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phylogeny , Protein Conformation , RNA, Small Interfering/genetics , Sequence Homology, Amino Acid
18.
Dev Cell ; 25(6): 553-4, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23806615

ABSTRACT

Reporting recently in Nature and Nature Structural and Molecular Biology, Shen et al. (2013) and Smibert et al. (2013) uncover mechanisms for Argonaute (Ago) protein regulation. Smibert et al. find that microRNA availability controls Ago levels, and Shen et al. show that epidermal growth factor receptor-mediated Ago2 phosphorylation affects Ago loading.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Cell Hypoxia/physiology , ErbB Receptors/metabolism , MicroRNAs/metabolism , Female , Humans
19.
Nat Struct Mol Biol ; 20(7): 814-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23665583

ABSTRACT

Argonaute proteins interact with small RNAs that guide them to complementary target RNAs, thus leading to inhibition of gene expression. Some but not all Argonaute proteins are endonucleases and can cleave the complementary target RNA. Here, we have mutated inactive human Ago1 and Ago3 and generated catalytic Argonaute proteins. We find that two short sequence elements at the N terminus are important for activity. In addition, PIWI-domain mutations in Ago1 may misarrange the catalytic center. Our work helps in understanding of the structural requirements that make an Argonaute protein an active endonucleolytic enzyme.


Subject(s)
Argonaute Proteins/chemistry , Eukaryotic Initiation Factors/chemistry , Amino Acid Sequence , Amino Acid Substitution , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Catalysis , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Humans , MicroRNAs/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Protein Conformation , Protein Structure, Tertiary , RNA Interference , RNA Processing, Post-Transcriptional , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
20.
J Cell Sci ; 126(Pt 1): 196-208, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23203810

ABSTRACT

During development, general body plan information must be translated into distinct morphologies of individual cells. Shaping cells is thought to involve cortical cytoskeletal components and Bin-Amphiphysin-Rvs167 (BAR) superfamily proteins. We therefore conducted comprehensive side-by-side loss-of-function studies of zebrafish orthologs of the F-BAR protein syndapin I and the actin nucleator Cobl. Zebrafish syndapin I associates with Cobl. The loss-of-function phenotypes of these proteins were remarkably similar and suggested a common function. Both cobl- and syndapin I-morphant fish showed severe swimming and balance-keeping defects, reflecting an impaired organization and function of the lateral line organ. Their lateral line organs lacked several neuromasts and showed an impaired functionality of the sensory hair cells within the neuromasts. Scanning electron microscopy revealed that sensory hair cells of both cobl- and syndapin I-morphant animals showed defects in the formation of both microtubule-dependent kinocilia and F-actin-rich stereocilia. Consistent with the kinocilia defects in sensory hair cells, body length was shortened and the development of body laterality, a process depending on motile cilia, was also impaired. Interestingly, Cobl and syndapin I both localized to the base of forming cilia. Rescue experiments demonstrated that proper formation of ciliated sensory hair cell rosettes relied on Cobl's syndapin I-binding Cobl homology domain, the actin-nucleating C-terminus of Cobl and the membrane curvature-inducing F-BAR domain of syndapin I. Our data thus suggest that the formation of distinct types of ciliary structures relies on membrane topology-modulating mechanisms that are based on F-BAR domain functions and on complex formation of syndapin I with the actin nucleator Cobl.


Subject(s)
Carrier Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Stereocilia/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cilia/metabolism , Cilia/ultrastructure , Humans , Immunohistochemistry , In Situ Hybridization , Microfilament Proteins/genetics , Microscopy, Atomic Force , Microscopy, Confocal , Stereocilia/ultrastructure , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...