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










Publication year range
1.
Reprod Toxicol ; 117: 108340, 2023 04.
Article in English | MEDLINE | ID: mdl-36731640

ABSTRACT

The role of spermatogonial stem cells (SSCs) is crucial in spermatogenesis, and extracellular vesicles (EVs) have been the focus of research as an important intercellular communication mechanism. Various endogenous regulatory factors secreted by Sertoli cells (SCs) can affect the self-maintenance and regeneration of SSCs, but little is known about the roles of SCs-derived exosomal microRNAs (miRNAs) on SSCs. In this study, we aimed to explore the regulation of the SCs-derived exosomal miR-30a-5p on SSCs proliferation and differentiation. EVs from the SCs were detected by electron microscopy and nanoparticle tracking analysis (NTA). Subsequently, the SSCs were treated with the SCs-derived extracellular vesicles (SCs-EVs). CCK-8 assay and EdU staining was applied to detect the cell proliferation, and the results indicated that SCs-EVs promoted the SSCs proliferation. Western blot detection of the SSCs markers (Gfrα1, Plzf, Stra8, and C-kit) indicated that SCs-EVs promoted the SSCs differentiation. Additionally, we found that SCs-EVs secreted miR-30a-5p to show the promoting effects. Besides, we discovered that miR-30a-5p targeted zinc finger E-box binding homeobox 2 (Zeb2) to regulate the ubiquitination of fibroblast growth factor 9 (Fgf9) in SSCs. miR-30a-3p/Zeb2/Fgf9 promoted the SSCs proliferation and differentiation by activating the mitogen­activated protein kinase (MAPK) signaling pathway. Taken together, our study showed that SCs-EVs can transport miR-30a-5p to SSCs and affect SSCs proliferation and differentiation by regulating the MAPK signaling pathway via Zeb2/Fgf9. This paper disclosed a novel molecular mechanism that regulates SSCs proliferation and differentiation, which could be valuable for the treatment of male infertility.


Subject(s)
MicroRNAs , Ubiquitin-Protein Ligases , Humans , Male , Sertoli Cells/metabolism , Ubiquitin , MicroRNAs/genetics , Cell Differentiation , Cell Proliferation , Stem Cells
2.
J Mater Chem B ; 11(11): 2431-2439, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36810648

ABSTRACT

Fluorescence imaging has enabled much progress in biological fields, while the evolution of commercially available dyes has lagged behind their advanced applications. Herein, we launch triphenylamine-equipped 1,8-naphthaolactam (NP-TPA) as a versatile scaffold for the custom design of an efficient subcellular imaging agent (NP-TPA-Tar), given its bright and constant emissions in various states, significant Stokes shifts, and facile modifiability. The resultant four NP-TPA-Tars maintain excellent emission behavior with targeted modifications and can map the spatial distribution of lysosomes, mitochondria, endoplasmic reticulum, and plasma membrane in Hep G2 cells. Compared to its commercial counterpart, NP-TPA-Tar has a 2.8-25.2 fold increase in Stokes shift, a 1.2-1.9 fold increase in photostability, enhanced targeting capability, and comparable imaging efficiency even at low concentrations of 50 nM. This work will help to accelerate the update of current imaging agents and super-resolution and real-time imaging in biological applications.


Subject(s)
Mitochondria , Optical Imaging , Amines
3.
PLoS One ; 16(4): e0248903, 2021.
Article in English | MEDLINE | ID: mdl-33857179

ABSTRACT

Following curative immunotherapy of B16F10 tumors, ~60% of mice develop a strong antibody response against cell-surface tumor antigens. Their antisera confer prophylactic protection against intravenous challenge with B16F10 cells, and also cross-react with syngeneic and allogeneic tumor cell lines MC38, EL.4, 4T1, and CT26. We identified the envelope glycoprotein (env) of a murine endogenous retrovirus (ERV) as the antigen accounting for the majority of this humoral response. A systemically administered anti-env monoclonal antibody cloned from such a response protects against tumor challenge, and prophylactic vaccination against the env protein protects a majority of naive mice from tumor establishment following subcutaneous inoculation with B16F10 cells. These results suggest the potential for effective prophylactic vaccination against analogous HERV-K env expressed in numerous human cancers.


Subject(s)
Antibodies, Neoplasm/immunology , Endogenous Retroviruses/immunology , Gene Products, env/immunology , Immunotherapy/methods , Neoplasms , Animals , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy
4.
Adv Mater ; 33(7): e2006057, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33448062

ABSTRACT

Short-wave infrared (SWIR, 900-1700 nm) enables in vivo imaging with high spatiotemporal resolution and penetration depth due to the reduced tissue autofluorescence and decreased photon scattering at long wavelengths. Although small organic SWIR dye molecules have excellent biocompatibility, they have been rarely exploited as compared to their inorganic counterparts, mainly due to their low quantum yield. To increase their brightness, in this work, the SWIR dye molecules are placed in close proximity to gold nanorods (AuNRs) for surface plasmon-enhanced emission. The fluorescence enhancement is optimized by controlling the dye-to-AuNR number ratio and up to ≈45-fold enhancement factor is achieved. In addition, the results indicate that the highest dye-to-AuNR number ratio gives the highest emission intensity per weight and this is used for synthesizing SWIR imaging probes using layer-by-layer (LbL) technique with polymer coating protection. Then, the SWIR imaging probes are applied for in vivo imaging of ovarian cancer and the surface coating effect on intratumor distribution of the imaging probes is investigated in two orthotopic ovarian cancer models. Lastly, it is demonstrated that the plasmon-enhanced SWIR imaging probe has great potential for fluorescence imaging-guided surgery by showing its capability to detect sub-millimeter-sized tumors.


Subject(s)
Biocompatible Materials/chemistry , Fluorescent Dyes/chemistry , Gold/chemistry , Nanotubes/chemistry , Optical Imaging/methods , Ovarian Neoplasms/diagnostic imaging , Animals , Biomedical Enhancement , Cell Line, Tumor , Female , Humans , Infrared Rays , Luciferases/chemistry , Luciferases/genetics , Mice, Nude , Polymers/chemistry , Radio Waves , Surface Properties , Tissue Distribution
6.
Nat Biomed Eng ; 5(5): 399-413, 2021 05.
Article in English | MEDLINE | ID: mdl-33046866

ABSTRACT

Therapies employing chimeric antigen receptor T cells (CAR-T cells) targeting tumour-associated antigens (TAAs) can lead to on-target-off-tumour toxicity and to resistance, owing to TAA expression in normal tissues and to TAA expression loss in tumour cells. These drawbacks can be circumvented by CAR-T cells targeting tumour-specific driver gene mutations, such as the four-nucleotide duplication in the oncogene nucleophosmin (NPM1c), which creates a neoepitope presented by the human leukocyte antigen with the A2 serotype (HLA-A2) that has been observed in about 35% of patients with acute myeloid leukaemia (AML). Here, we report a human single-chain variable fragment (scFv), identified via yeast surface display, that specifically binds to the NPM1c epitope-HLA-A2 complex but not to HLA-A2 or to HLA-A2 loaded with control peptides. In vitro and in mice, CAR-T cells with the scFv exhibit potent cytotoxicity against NPM1c+HLA-A2+ leukaemia cells and primary AML blasts, but not NPM1c-HLA-A2+ leukaemia cells or HLA-A2- tumour cells. Therapies using NPM1c CAR-T cells for the treatment of NPM1c+HLA-A2+ AML may limit on-target-off-tumour toxicity and tumour resistance.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Leukemia, Myeloid, Acute/therapy , Nuclear Proteins/chemistry , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/administration & dosage , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Epitopes/immunology , HLA-A2 Antigen/immunology , Humans , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/immunology , Mice , Nuclear Proteins/immunology , Nucleophosmin , PC-3 Cells , Proof of Concept Study , Single-Chain Antibodies/pharmacology
7.
Nat Commun ; 11(1): 4837, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973183

ABSTRACT

ATP synthesis and thermogenesis are two critical outputs of mitochondrial respiration. How these outputs are regulated to balance the cellular requirement for energy and heat is largely unknown. Here we show that major facilitator superfamily domain containing 7C (MFSD7C) uncouples mitochondrial respiration to switch ATP synthesis to thermogenesis in response to heme. When heme levels are low, MSFD7C promotes ATP synthesis by interacting with components of the electron transport chain (ETC) complexes III, IV, and V, and destabilizing sarcoendoplasmic reticulum Ca2+-ATPase 2b (SERCA2b). Upon heme binding to the N-terminal domain, MFSD7C dissociates from ETC components and SERCA2b, resulting in SERCA2b stabilization and thermogenesis. The heme-regulated switch between ATP synthesis and thermogenesis enables cells to match outputs of mitochondrial respiration to their metabolic state and nutrient supply, and represents a cell intrinsic mechanism to regulate mitochondrial energy metabolism.


Subject(s)
Adenosine Triphosphate/metabolism , Heme/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Receptors, Virus/metabolism , Thermogenesis/physiology , Animals , Cytochrome-c Oxidase Deficiency , Electron Transport Complex III , Electron Transport Complex IV , Energy Metabolism/physiology , Gene Knockout Techniques , HEK293 Cells , Humans , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Protein Domains , Receptors, Virus/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , THP-1 Cells
8.
Sci Adv ; 6(24): eaba7589, 2020 06.
Article in English | MEDLINE | ID: mdl-32582856

ABSTRACT

The stimulator of interferon (IFN) genes (STING) pathway constitutes a highly important part of immune responses against various cancers and infections. Consequently, administration of STING agonists such as cyclic GMP-AMP (cGAMP) has been identified as a promising approach to target these diseases. In cancer cells, STING signaling is frequently impaired by epigenetic silencing of STING; hence, conventional delivery of only its agonist cGAMP may be insufficient to trigger STING signaling. In this work, while expression of STING lacking the transmembrane (TM) domain is known to be unresponsive to STING agonists and is dominant negative when coexpressed with the full-length STING inside cells, we observed that the recombinant TM-deficient STING protein complexed with cGAMP could effectively trigger STING signaling when delivered in vitro and in vivo, including in STING-deficient cell lines. Thus, this bioinspired method using TM-deficient STING may present a universally applicable platform for cGAMP delivery.


Subject(s)
Nucleotides, Cyclic , Signal Transduction , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism
9.
Nat Cancer ; 1(9): 882-893, 2020 09.
Article in English | MEDLINE | ID: mdl-34447945

ABSTRACT

Therapies that synergistically stimulate immunogenic cancer cell death (ICD), inflammation, and immune priming are of great interest for cancer immunotherapy. However, even multi-agent therapies often fail to trigger all of the steps necessary for self-sustaining anti-tumor immunity. Here we describe self-replicating RNAs encapsulated in lipid nanoparticles (LNP-replicons), which combine three key elements: (1) an LNP composition that potently promotes ICD, (2) RNA that stimulates danger sensors in transfected cells, and (3) RNA-encoded IL-12 for modulation of immune cells. Intratumoral administration of LNP-replicons led to high-level expression of IL-12, stimulation of a type I interferon response, and cancer cell ICD, resulting in a highly inflamed tumor microenvironment and priming of systemic anti-tumor immunity. In several mouse models of cancer, a single intratumoral injection of replicon-LNPs eradicated large established tumors, induced protective immune memory, and enabled regression of distal uninjected tumors. LNP-replicons are thus a promising multifunctional single-agent immunotherapeutic.


Subject(s)
Nanoparticles , Neoplasms , Animals , Immunotherapy/methods , Interleukin-12/genetics , Liposomes , Mice , Neoplasms/genetics , RNA , Tumor Microenvironment
10.
Sci Rep ; 9(1): 6932, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31061426

ABSTRACT

Self-replicating (replicon) RNA is a promising new platform for gene therapy, but applications are still limited by short persistence of expression in most cell types and low levels of transgene expression in vivo. To address these shortcomings, we developed an in vitro evolution strategy and identified six mutations in nonstructural proteins (nsPs) of Venezuelan equine encephalitis (VEE) replicon that promoted subgenome expression in cells. Two mutations in nsP2 and nsP3 enhanced transgene expression, while three mutations in nsP3 regulated this expression. Replicons containing the most effective mutation combinations showed enhanced duration and cargo gene expression in vivo. In comparison to wildtype replicon, mutants expressing IL-2 injected into murine B16F10 melanoma showed 5.5-fold increase in intratumoral IL-2 and 2.1-fold increase in infiltrating CD8 T cells, resulting in significantly slowed tumor growth. Thus, these mutant replicons may be useful for improving RNA therapeutics for vaccination, cancer immunotherapy, and gene therapy.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Immunotherapy , RNA/genetics , Replicon , Transcription, Genetic , Alleles , Animals , Cell Line, Tumor , Gene Expression , Gene Order , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Immunity/genetics , Immunotherapy/methods , Melanoma, Experimental , Mice , Mutation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Promoter Regions, Genetic , RNA/administration & dosage
11.
ACS Nano ; 12(10): 10272-10280, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30272942

ABSTRACT

The utility of layer-by-layer (LbL) coated microneedle (MN) skin patches for transdermal drug delivery has proven to be a promising approach, with advantages over hypodermal injection due to painless and easy self-administration. However, the long epidermal application time required for drug implantation by existing LbL MN strategies (15-90 min) can lead to potential medication noncompliance. Here, we developed a MN platform to shorten the application time in MN therapies based on a synthetic pH-induced charge-invertible polymer poly(2-(diisopropylamino) ethyl methacrylate- b-methacrylic acid) (PDM), requiring only 1 min skin insertion time to implant LbL films in vivo. Following MN-mediated delivery of 0.5 µg model antigen chicken ovalbumin (OVA) in the skin of mice, this system achieved sustained release over 3 days and led to an elevated immune response as demonstrated by significantly higher humoral immunity compared with OVA administration via conventional routes (subcutaneously and intramuscularly). Moreover, in an ex vivo experiment on human skin, we achieved efficient immune activation through MN-delivered LbL films, demonstrated by a rapid uptake of vaccine adjuvants by the antigen presenting cells. These features, rapid administration and the ability to elicit a robust immune response, can potentially enable a broad application of microneedle-based vaccination technologies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Needles , Oligodeoxyribonucleotides/pharmacology , Polymethacrylic Acids/chemical synthesis , Programmed Cell Death 1 Receptor/immunology , Skin/drug effects , Adjuvants, Immunologic/administration & dosage , Administration, Cutaneous , Animals , Chickens , Drug Delivery Systems , Female , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polymethacrylic Acids/chemistry , Skin/immunology , Vaccination
12.
ACS Nano ; 11(3): 2531-2544, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28157292

ABSTRACT

Messenger RNA (mRNA) represents a promising class of nucleic-acid-based therapeutics. While numerous nanocarriers have been developed for mRNA delivery, the inherent labile nature of mRNA results in a very low transfection efficiency and poor expression of desired protein. Here we preassemble the mRNA translation initiation structure through an inherent molecular recognition between 7-methylguanosine (m7G)-capped mRNA and eukaryotic initiation factor 4E (eIF4E) protein to form ribonucleoproteins (RNPs), thereby mimicking the first step of protein synthesis inside cells. Subsequent electrostatic stabilization of RNPs with structurally tunable cationic carriers leads to nanosized complexes (nanoplexes), which elicit high levels of mRNA transfection in different cell types by enhancing intracellular mRNA stability and protein synthesis. By investigating a family of synthetic polypeptides bearing different side group arrangements of cationic charge, we find that the molecular structure modulates the nanoscale distance between the mRNA strand and the eIF4E protein inside the nanoplex, which directly impacts the enhancement of mRNA transfection. To demonstrate the biomedical potential of this approach, we use this approach to introduce mRNA/eIF4E nanoplexes to murine dendritic cells, resulting in increased activation of cytotoxic CD8 T cells ex vivo. More importantly, eIF4E enhances gene expression in lungs following a systemic delivery of luciferase mRNA/eIF4E in mice. Collectively, this bioinspired molecular assembly method could lead to a new paradigm of gene delivery.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Gene Transfer Techniques , Nanoparticles/chemistry , Peptide Chain Initiation, Translational/genetics , RNA Caps/genetics , RNA, Messenger/genetics , Eukaryotic Initiation Factor-4E/chemistry , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/chemistry
13.
J Immunol ; 195(4): 1591-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163589

ABSTRACT

Recall responses by memory CD8 T cells are impaired in the absence of CD4 T cells. Although several mechanisms have been proposed, the molecular basis is still largely unknown. Using a local influenza virus infection in the respiratory tract and the lung of CD4(-/-) mice, we show that memory CD8 T cell impairment is limited to the lungs and the lung-draining lymph nodes, where viral Ags are unusually persistent and abundant in these mice. Persistent Ag exposure results in prolonged activation of the AKT-mTORC1 pathway in Ag-specific CD8 T cells, favoring their development into effector memory T cells at the expense of central memory T cells, and inhibition of mTORC1 by rapamycin largely corrects the impairment by promoting central memory T cell development. The findings suggest that the prolonged AKT-mTORC1 activation driven by persistent Ag is a critical mechanism underlying the impaired memory CD8 T cell development and responses in the absence of CD4 T cells.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Immunophenotyping , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Phenotype , Sirolimus/pharmacology
14.
PLoS One ; 8(7): e67590, 2013.
Article in English | MEDLINE | ID: mdl-23844039

ABSTRACT

We present a method for direct non-optical quantification of dry mass, dry density and water mass of single living cells in suspension. Dry mass and dry density are obtained simultaneously by measuring a cell's buoyant mass sequentially in an H2O-based fluid and a D2O-based fluid. Rapid exchange of intracellular H2O for D2O renders the cell's water content neutrally buoyant in both measurements, and thus the paired measurements yield the mass and density of the cell's dry material alone. Utilizing this same property of rapid water exchange, we also demonstrate the quantification of intracellular water mass. In a population of E. coli, we paired these measurements to estimate the percent dry weight by mass and volume. We then focused on cellular dry density - the average density of all cellular biomolecules, weighted by their relative abundances. Given that densities vary across biomolecule types (RNA, DNA, protein), we investigated whether we could detect changes in biomolecular composition in bacteria, fungi, and mammalian cells. In E. coli, and S. cerevisiae, dry density increases from stationary to exponential phase, consistent with previously known increases in the RNA/protein ratio from up-regulated ribosome production. For mammalian cells, changes in growth conditions cause substantial shifts in dry density, suggesting concurrent changes in the protein, nucleic acid and lipid content of the cell.


Subject(s)
DNA/analysis , Lipids/analysis , Proteins/analysis , RNA/analysis , Water/metabolism , Animals , Biological Transport , Deuterium Exchange Measurement , Erythrocytes/chemistry , Escherichia coli/chemistry , Fibroblasts/chemistry , Humans , Mice , Saccharomyces cerevisiae/chemistry , T-Lymphocytes/chemistry
15.
Plant Cell ; 24(5): 2225-36, 2012 May.
Article in English | MEDLINE | ID: mdl-22643122

ABSTRACT

In Arabidopsis thaliana, the MEKK1-MKK1/MKK2-MPK4 mitogen-activated protein (MAP) kinase cascade represses cell death and immune responses. In mekk1, mkk1 mkk2, and mpk4 mutants, programmed cell death and defense responses are constitutively activated, but the mechanism by which MEKK1, MKK1/MKK2, and MPK4 negatively regulate cell death and immunity was unknown. From a screen for suppressors of mkk1 mkk2, we found that mutations in suppressor of mkk1 mkk2 1 (summ1) suppress the cell death and defense responses not only in mkk1 mkk2 but also in mekk1 and mpk4. SUMM1 encodes the MAP kinase kinase kinase MEKK2. It interacts with MPK4 and is phosphorylated by MPK4 in vitro. Overexpression of SUMM1 activates cell death and defense responses that are dependent on the nucleotide binding-leucine-rich repeat protein SUMM2. Taken together, our data suggest that the MEKK1-MKK1/MKK2-MPK4 kinase cascade negatively regulates MEKK2 and activation of MEKK2 triggers SUMM2-mediated immune responses.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/immunology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Phosphorylation , Plant Immunity/genetics , Plant Immunity/physiology
16.
Proc Natl Acad Sci U S A ; 108(35): 14694-9, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21873230

ABSTRACT

The nucleotide-binding domain and leucine-rich repeats containing proteins (NLRs) serve as immune receptors in both plants and animals. Overaccumulation of NLRs often leads to autoimmune responses, suggesting that the levels of these immune receptors must be tightly controlled. However, the mechanism by which NLR protein levels are regulated is unknown. Here we report that the F-box protein CPR1 controls the stability of plant NLR resistance proteins. Loss-of-function mutations in CPR1 lead to higher accumulation of the NLR proteins SNC1 and RPS2, as well as autoactivation of immune responses. The autoimmune responses in cpr1 mutant plants can be largely suppressed by knocking out SNC1. Furthermore, CPR1 interacts with SNC1 and RPS2 in vivo, and overexpressing CPR1 results in reduced accumulation of SNC1 and RPS2, as well as suppression of immunity mediated by these two NLR proteins. Our data suggest that SKP1-CULLIN1-F-box (SCF) complex-mediated stability control of plant NLR proteins plays an important role in regulating their protein levels and preventing autoimmunity.


Subject(s)
Arabidopsis Proteins/physiology , Immunity, Innate , Plants/immunology , S-Phase Kinase-Associated Proteins/physiology , Autoimmunity , F-Box Proteins/physiology , Mutation , Phenotype , Plants/metabolism
17.
Plant Signal Behav ; 6(3): 434-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21350329

ABSTRACT

MOS1 (MODIFIER OF snc1) was identified through a genetic screen for suppressors of snc1, an autoimmune mutant caused by a gain-of-function mutation in a TIR-NB-LRR-type Resistance gene. Loss of MOS1 function completely suppresses snc1-mediated autoimmunity. The MOS1 protein contains a BAT2 domain and regulates the expression of SNC1 in a locus-specific manner, but the mechanism on how MOS1 epigenetically regulates SNC1 gene expression is unclear. Here, we report the gene expression pattern and subcellular localization of MOS1. In addition, we analyze and discuss the roles of DNA and histone methylation in mos1-mediated suppression of SNC1 expression.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Transcription Factors/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
18.
PLoS Pathog ; 6(9): e1001111, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20862316

ABSTRACT

Plant defense responses need to be tightly regulated to prevent auto-immunity, which is detrimental to growth and development. To identify negative regulators of Resistance (R) protein-mediated resistance, we screened for mutants with constitutive defense responses in the npr1-1 background. Map-based cloning revealed that one of the mutant genes encodes a conserved TPR domain-containing protein previously known as SRFR1 (SUPPRESSOR OF rps4-RLD). The constitutive defense responses in the srfr1 mutants in Col-0 background are suppressed by mutations in SNC1, which encodes a TIR-NB-LRR (Toll Interleukin1 Receptor-Nucleotide Binding-Leu-Rich Repeat) R protein. Yeast two-hybrid screens identified SGT1a and SGT1b as interacting proteins of SRFR1. The interactions between SGT1 and SRFR1 were further confirmed by co-immunoprecipitation analysis. In srfr1 mutants, levels of multiple NB-LRR R proteins including SNC1, RPS2 and RPS4 are increased. Increased accumulation of SNC1 is also observed in the sgt1b mutant. Our data suggest that SRFR1 functions together with SGT1 to negatively regulate R protein accumulation, which is required for preventing auto-activation of plant immunity.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Glucosyltransferases/metabolism , Plant Immunity/physiology , Plant Proteins/genetics , Arabidopsis/genetics , Arabidopsis/growth & development , Blotting, Western , Gene Expression Regulation, Plant , Glucosyltransferases/genetics , Immunoprecipitation , Mutation/genetics , Plant Diseases/genetics , Plant Diseases/immunology , Plant Proteins/metabolism , RNA, Messenger/genetics , RNA, Plant/genetics , Two-Hybrid System Techniques
19.
Plant Physiol ; 153(3): 1425-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439546

ABSTRACT

Plant Resistance (R) genes encode immune receptors that recognize pathogens and activate defense responses. Because of fitness costs associated with maintaining R protein-mediated resistance, expression levels of R genes have to be tightly regulated. However, mechanisms on how R-gene expression is regulated are poorly understood. Here we show that MODIFIER OF snc1, 1 (MOS1) regulates the expression of SUPPRESSOR OF npr1-1, CONSTITUTIVE1 (SNC1), which encodes a Toll/interleukin receptor-nucleotide binding site-leucine-rich repeat type of R protein in Arabidopsis (Arabidopsis thaliana). In the mos1 loss-of-function mutant plants, snc1 expression is repressed and constitutive resistance responses mediated by snc1 are lost. The repression of snc1 expression in mos1 is released by knocking out DECREASE IN DNA METHYLATION1. In mos1 mutants, DNA methylation in a region upstream of SNC1 is altered. Furthermore, expression of snc1 transgenes using the native promoter does not require MOS1, indicating that regulation of SNC1 expression by MOS1 is at the chromatin level. Map-based cloning of MOS1 revealed that it encodes a novel protein with a HLA-B ASSOCIATED TRANSCRIPT2 (BAT2) domain that is conserved in plants and animals. Our study on MOS1 suggests that BAT2 domain-containing proteins may function in regulation of gene expression at chromatin level.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Conserved Sequence/genetics , Gene Expression Regulation, Plant , Immunity, Innate/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Alleles , Amino Acid Sequence , Animals , Arabidopsis/microbiology , Arabidopsis Proteins/genetics , Chromatin/metabolism , Cloning, Molecular , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Plant/genetics , Genetic Loci , Molecular Sequence Data , Mutation/genetics , Phenotype , Plant Diseases/genetics , Plant Diseases/immunology , Plant Diseases/microbiology , Plants, Genetically Modified , Protein Structure, Tertiary , Sequence Alignment , Transcription Factors/genetics
20.
PLoS Genet ; 6(12): e1001250, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21203492

ABSTRACT

Nucleocytoplasmic trafficking is emerging as an important aspect of plant immunity. The three related pathways affecting plant immunity include Nuclear Localization Signal (NLS)-mediated nuclear protein import, Nuclear Export Signal (NES)-dependent nuclear protein export, and mRNA export relying on MOS3, a nucleoporin belonging to the Nup107-160 complex. Here we report the characterization, identification, and detailed analysis of Arabidopsis modifier of snc1, 11 (mos11). Mutations in MOS11 can partially suppress the dwarfism and enhanced disease resistance phenotypes of snc1, which carries a gain-of-function mutation in a TIR-NB-LRR type Resistance gene. MOS11 encodes a conserved eukaryotic protein with homology to the human RNA binding protein CIP29. Further functional analysis shows that MOS11 localizes to the nucleus and that the mos11 mutants accumulate more poly(A) mRNAs in the nucleus, likely resulting from reduced mRNA export activity. Epistasis analysis between mos3-1 and mos11-1 revealed that MOS11 probably functions in the same mRNA export pathway as MOS3, in a partially overlapping fashion, before the mRNA molecules pass through the nuclear pores. Taken together, MOS11 is identified as a new protein contributing to the transfer of mature mRNA from the nucleus to the cytosol.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cell Nucleus/metabolism , RNA, Messenger/metabolism , RNA, Plant/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Biological Transport , Cell Nucleus/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , RNA, Messenger/genetics , RNA, Plant/genetics , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...