Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Immunol ; 20(6): 747-755, 2019 06.
Article in English | MEDLINE | ID: mdl-31061531

ABSTRACT

Despite gathering evidence that ubiquitylation can direct non-degradative outcomes, most investigations of ubiquitylation in T cells have focused on degradation. Here, we integrated proteomic and transcriptomic datasets from primary mouse CD4+ T cells to establish a framework for predicting degradative or non-degradative outcomes of ubiquitylation. Di-glycine remnant profiling was used to reveal ubiquitylated proteins, which in combination with whole-cell proteomic and transcriptomic data allowed prediction of protein degradation. Analysis of ubiquitylated proteins identified by di-glycine remnant profiling indicated that activation of CD4+ T cells led to an increase in non-degradative ubiquitylation. This correlated with an increase in non-proteasome-targeted K29, K33 and K63 polyubiquitin chains. This study revealed over 1,200 proteins that were ubiquitylated in primary mouse CD4+ T cells and highlighted the relevance of non-proteasomally targeted ubiquitin chains in T cell signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , Proteome , Proteomics , Animals , Gene Expression Profiling , Lymphocyte Activation/genetics , Mass Spectrometry , Mice , Polyubiquitin/metabolism , Proteomics/methods , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Ubiquitination
2.
Cell ; 157(7): 1577-90, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949970

ABSTRACT

Clec16a has been identified as a disease susceptibility gene for type 1 diabetes, multiple sclerosis, and adrenal dysfunction, but its function is unknown. Here we report that Clec16a is a membrane-associated endosomal protein that interacts with E3 ubiquitin ligase Nrdp1. Loss of Clec16a leads to an increase in the Nrdp1 target Parkin, a master regulator of mitophagy. Islets from mice with pancreas-specific deletion of Clec16a have abnormal mitochondria with reduced oxygen consumption and ATP concentration, both of which are required for normal ß cell function. Indeed, pancreatic Clec16a is required for normal glucose-stimulated insulin release. Moreover, patients harboring a diabetogenic SNP in the Clec16a gene have reduced islet Clec16a expression and reduced insulin secretion. Thus, Clec16a controls ß cell function and prevents diabetes by controlling mitophagy. This pathway could be targeted for prevention and control of diabetes and may extend to the pathogenesis of other Clec16a- and Parkin-associated diseases.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Islets of Langerhans/pathology , Lectins, C-Type/metabolism , Mitophagy , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Diabetes Mellitus, Type 1/pathology , Genetic Predisposition to Disease , Glucose/metabolism , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lysosomes/chemistry , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Polymorphism, Single Nucleotide , Ubiquitin-Protein Ligases
3.
Blood ; 142(20): 1708-1723, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37699202

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) transplantation serves as a curative therapy for many benign and malignant hematopoietic disorders and as a platform for gene therapy. However, growing needs for ex vivo manipulation of HSPC-graft products are limited by barriers in maintaining critical self-renewal and quiescence properties. The role of sphingolipid metabolism in safeguarding these essential cellular properties has been recently recognized, but not yet widely explored. Here, we demonstrate that pharmacologic and genetic inhibition of neutral sphingomyelinase 2 (nSMase-2) leads to sustained improvements in long-term competitive transplantation efficiency after ex vivo culture. Mechanistically, nSMase-2 blockade activates a canonical integrated stress response (ISR) and promotes metabolic quiescence in human and murine HSPCs. These adaptations result in part from disruption in sphingolipid metabolism that impairs the release of nSMase-2-dependent extracellular vesicles (EVs). The aggregate findings link EV trafficking and the ISR as a regulatory dyad guarding HSPC homeostasis and long-term fitness. Translationally, transient nSMase-2 inhibition enables ex vivo graft manipulation with enhanced HSPC potency.


Subject(s)
Hematopoietic Stem Cell Transplantation , Sphingomyelin Phosphodiesterase , Animals , Humans , Mice , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Hematopoietic Stem Cells/metabolism , Sphingolipids/metabolism
4.
PLoS Biol ; 19(2): e3001041, 2021 02.
Article in English | MEDLINE | ID: mdl-33524014

ABSTRACT

The capacity for T cells to become activated and clonally expand during pathogen invasion is pivotal for protective immunity. Our understanding of how T cell receptor (TCR) signaling prepares cells for this rapid expansion remains limited. Here we provide evidence that the E3 ubiquitin ligase Cullin-4b (Cul4b) regulates this process. The abundance of total and neddylated Cul4b increased following TCR stimulation. Disruption of Cul4b resulted in impaired proliferation and survival of activated T cells. Additionally, Cul4b-deficient CD4+ T cells accumulated DNA damage. In T cells, Cul4b preferentially associated with the substrate receptor DCAF1, and Cul4b and DCAF1 were found to interact with proteins that promote the sensing or repair of damaged DNA. While Cul4b-deficient CD4+ T cells showed evidence of DNA damage sensing, downstream phosphorylation of SMC1A did not occur. These findings reveal an essential role for Cul4b in promoting the repair of damaged DNA to allow survival and expansion of activated T cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , DNA Repair/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/metabolism , Cell Proliferation/physiology , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA Damage , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell , Signal Transduction , Ubiquitin-Protein Ligases/genetics
5.
Mol Genet Metab ; 138(2): 107371, 2023 02.
Article in English | MEDLINE | ID: mdl-36709534

ABSTRACT

Mucopolysaccharidosis I is a lysosomal storage disorder characterized by deficient alpha-L-iduronidase activity, leading to abnormal accumulation of glycosaminoglycans in cells and tissues. Synovial joint disease is prevalent and significantly reduces patient quality of life. There is a critical need for improved understanding of joint disease pathophysiology in MPS I, including specific biomarkers to predict and monitor joint disease progression, and response to treatment. The objective of this study was to leverage the naturally-occurring MPS I canine model and undertake an unbiased proteomic screen to identify systemic biomarkers predictive of local joint disease in MPS I. Synovial fluid and serum samples were collected from MPS I and healthy dogs at 12 months-of-age, and protein abundance characterized using liquid chromatography tandem mass spectrometry. Stifle joints were evaluated postmortem using magnetic resonance imaging (MRI) and histology. Proteomics identified 40 proteins for which abundance was significantly correlated between serum and synovial fluid, including markers of inflammatory joint disease and lysosomal dysfunction. Elevated expression of three biomarker candidates, matrix metalloproteinase 19, inter-alpha-trypsin inhibitor heavy-chain 3 and alpha-1-microglobulin, was confirmed in MPS I cartilage, and serum abundance of these molecules was found to correlate with MRI and histological degenerative grades. The candidate biomarkers identified have the potential to improve patient care by facilitating minimally-invasive, specific assessment of joint disease progression and response to therapeutic intervention.


Subject(s)
Joint Diseases , Mucopolysaccharidosis I , Dogs , Animals , Mucopolysaccharidosis I/pathology , Proteomics , Quality of Life , Joint Diseases/metabolism , Synovial Fluid/metabolism , Biomarkers/metabolism , Disease Progression
6.
Blood ; 137(3): 392-397, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32959045

ABSTRACT

Neutrophils are critical mediators of host defense in pathogen-induced and sterile inflammation. Excessive neutrophil activation has been associated with increased host pathology through collateral organ damage. The beneficial aspects of neutrophil activation, particularly in sterile inflammation, are less well defined. We observed accumulation of nuclear debris in the lungs of neutropenic mice exposed to acid-induced injury compared with wild type. Size analysis of DNA debris showed that neutropenic mice were unable to degrade extracellular DNA fragments. In addition, we found that neutrophils are able to differentially express DNA-degrading and repair-associated genes and proteins. Once neutrophils are at sites of lung inflammation, they are able to phagocytose and degrade extracellular DNA. This neutrophil-dependent DNA degradation occurs in a MyD88-dependent pathway. The increased DNA debris in neutropenic mice was associated with dysregulated alveolar repair and the phenotype is rescued by intratracheal administration of DNase I. Thus, we show a novel mechanism as part of the inflammatory response, in which neutrophils engulf and degrade extracellular DNA fragments and allow for optimal organ repair.


Subject(s)
Acids/adverse effects , Cell Nucleus/pathology , Lung Injury/pathology , Neutrophils/pathology , Animals , Bronchoalveolar Lavage Fluid , DNA/metabolism , Extracellular Space/metabolism , Granulocyte Colony-Stimulating Factor/deficiency , Granulocyte Colony-Stimulating Factor/metabolism , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Neutropenia/pathology , Wound Healing
7.
Am J Physiol Lung Cell Mol Physiol ; 322(3): L365-L372, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34984927

ABSTRACT

Both sepsis and acute respiratory distress syndrome (ARDS) rely on imprecise clinical definitions leading to heterogeneity, which has contributed to negative trials. Because circulating protein/DNA complexes have been implicated in sepsis and ARDS, we aimed to develop a proteomic signature of DNA-bound proteins to discriminate between children with sepsis with and without ARDS. We performed a prospective case-control study in 12 children with sepsis with ARDS matched to 12 children with sepsis without ARDS on age, severity of illness score, and source of infection. We performed co-immunoprecipitation and downstream proteomics in plasma collected ≤ 24 h of intensive care unit admission. Expression profiles were generated, and a random forest classifier was used on differentially expressed proteins to develop a signature which discriminated ARDS. The classifier was tested in six independent blinded samples. Neutrophil and nucleosome proteins were over-represented in ARDS, including two S100A proteins, superoxide dismutase (SOD), and three histones. Random forest produced a 10-protein signature that accurately discriminated between children with sepsis with and without ARDS. This classifier perfectly assigned six independent blinded samples as having ARDS or not. We validated higher expression of the most informative discriminating protein, galectin-3-binding protein, in children with ARDS. Our methodology has applicability to isolation of DNA-bound proteins from plasma. Our results support the premise of a molecular definition of ARDS, and give preliminary insight into why some children with sepsis, but not others, develop ARDS.


Subject(s)
Respiratory Distress Syndrome , Sepsis , Case-Control Studies , Child , DNA , Humans , Proteomics , Respiratory Distress Syndrome/diagnosis , Sepsis/complications , Sepsis/diagnosis
8.
Eur J Immunol ; 50(10): 1468-1483, 2020 10.
Article in English | MEDLINE | ID: mdl-32459862

ABSTRACT

To mount an antipathogen response, CD4 T cells must undergo rapid cell proliferation; however, poorly controlled expansion can result in diseases such as autoimmunity. One important regulator of T-cell activity is the E3 ubiquitin ligase Itch. Itch deficient patients suffer from extensive autoinflammation. Similarly, Itch deficient mice exhibit inflammation characterized by high numbers of activated CD4 T cells. While the role of Itch in limiting CD4 T-cell cytokine production has been extensively studied, it is less clear whether and how Itch regulates proliferation of these cells. We determined that Itch deficient CD4 T cells are hyperproliferative in vitro and in vivo, due to increased S phase entry. Whole cell proteomics analysis of Itch deficient primary mouse CD4 T cells revealed increased abundance of the ß-catenin coactivator WW domain-binding protein 2 (WBP2). Furthermore, Itch deficient cells demonstrate increased WBP2 protein stability, and Itch and WBP2 interact in CD4 T cells. Knockdown of WBP2 in CD4 T cells caused reduced proliferation. Together, our data support that Itch attenuates CD4 T cell proliferation by promoting WBP2 degradation. This study identifies novel roles for Itch and WBP2 in regulating CD4 T cell proliferation, providing insight into how Itch may prevent inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Pruritus/immunology , Syk Kinase/metabolism , Trans-Activators/metabolism , Animals , Autoantigens/immunology , Autoimmunity , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Lymphocyte Activation , Mice , Protein Stability , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
9.
Crit Care Med ; 49(7): 1149-1158, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33729723

ABSTRACT

OBJECTIVES: Circulating nucleosomes and their component histones have been implicated as pathogenic in sepsis and acute respiratory distress syndrome in adults. However, their role in pediatric acute respiratory distress syndrome is unknown. DESIGN: We performed a prospective cohort study in children with acute respiratory distress syndrome, with plasma collection within 24 hours of acute respiratory distress syndrome onset. We associated nucleosome levels with severity of acute respiratory distress syndrome and with nonpulmonary organ failures and tested for association of nucleosomes with PICU mortality and ventilator-free days at 28 days in univariate and multivariable analyses. We also performed proteomics of DNA-bound plasma proteins in a matched case-control study of septic children with and without acute respiratory distress syndrome in order to identify specific histone proteins elevated in acute respiratory distress syndrome. SETTING: Large academic tertiary-care PICU. PATIENTS: Intubated children meeting Berlin criteria for acute respiratory distress syndrome. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We enrolled 333 children with acute respiratory distress syndrome, with 69 nonsurvivors (21%). Plasma nucleosomes were correlated with acute respiratory distress syndrome severity and with the number of nonpulmonary organ failures at acute respiratory distress syndrome onset. Nucleosomes were higher (p < 0.001) in nonsurvivors (0.40 [interquartile range, 0.20-0.71] arbitrary units) relative to survivors (0.10 [interquartile range, 0.04-0.25] arbitrary units). Nucleosomes were associated with PICU mortality in multivariable analysis (adjusted odds ratio 1.84 per 1 sd increase; 95% CI, 1.38-2.45; p < 0.001). Nucleosomes were also associated with a lower probability of being extubated alive by day 28 after multivariable adjustment (adjusted subdistribution hazard ratio, 0.74; 95% CI, 0.63-0.88; p = 0.001). Proteomic analysis demonstrated higher levels of the core nucleosome histones H2A, H2B, H3, and H4 in septic children with acute respiratory distress syndrome, relative to septic children without acute respiratory distress syndrome. CONCLUSIONS: Plasma nucleosomes are associated with acute respiratory distress syndrome severity, nonpulmonary organ failures, and worse outcomes in pediatric acute respiratory distress syndrome.


Subject(s)
Histones/blood , Nucleosomes/metabolism , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/mortality , Adolescent , Airway Extubation , Case-Control Studies , Child , Child, Preschool , DNA/blood , Female , Hospital Mortality , Humans , Intensive Care Units, Pediatric , Male , Multiple Organ Failure/mortality , Prognosis , Prospective Studies , Proteomics , Respiration, Artificial , Respiratory Distress Syndrome/complications , Sepsis/blood , Sepsis/complications , Severity of Illness Index , Survival Rate
10.
J Proteome Res ; 19(4): 1857-1862, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32129078

ABSTRACT

The growing field of urinary proteomics shows promise to expand the number of biomarkers for the diagnosis and prognosis of a number of human diseases. With the rapid developments in mass spectrometry methods for proteome quantification, there exists an opportunity for improved sample processing and separation workflows to make important contributions to urine proteomic analyses. Here we evaluate the performance of four sample preparation methods: MStern, PreOmics in-StageTip (iST), suspension-trapping (S-Trap), and conventional urea In-Solution trypsin hydrolysis for nondepleted urine samples. Data-dependent acquisition (DDA) mode on a QExactive HF mass spectrometer was used for single-shot label-free data acquisition. Our results demonstrate a high degree of reproducibility within each workflow. PreOmics iST yields the best digestion efficiency, whereas the S-Trap workflow gives the greatest number of peptide and protein identifications. Using the S-Trap method and starting with ∼0.5 mL, we identify ∼1500 protein groups and ∼17 700 peptides from DDA analysis with a single injection on the mass spectrometer.


Subject(s)
Proteome , Proteomics , Humans , Mass Spectrometry , Reproducibility of Results , Specimen Handling , Workflow
11.
Mol Cell Proteomics ; 16(12): 2079-2097, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28972080

ABSTRACT

Viral DNA genomes replicating in cells encounter a myriad of host factors that facilitate or hinder viral replication. Viral proteins expressed early during infection modulate host factors interacting with viral genomes, recruiting proteins to promote viral replication, and limiting access to antiviral repressors. Although some host factors manipulated by viruses have been identified, we have limited knowledge of pathways exploited during infection and how these differ between viruses. To identify cellular processes manipulated during viral replication, we defined proteomes associated with viral genomes during infection with adenovirus, herpes simplex virus and vaccinia virus. We compared enrichment of host factors between virus proteomes and confirmed association with viral genomes and replication compartments. Using adenovirus as an illustrative example, we uncovered host factors deactivated by early viral proteins, and identified a subgroup of nucleolar proteins that aid virus replication. Our data sets provide valuable resources of virus-host interactions that affect proteins on viral genomes.


Subject(s)
Dependovirus/physiology , Proteome/metabolism , Simplexvirus/physiology , Vaccinia virus/physiology , Viral Proteins/metabolism , Virus Diseases/metabolism , A549 Cells , Cell Line, Tumor , DNA Replication , Genome, Viral , HeLa Cells , Host-Pathogen Interactions , Humans , Protein Interaction Maps , Proteomics/methods , Virus Replication
12.
Proteomics ; 17(7)2017 04.
Article in English | MEDLINE | ID: mdl-28116781

ABSTRACT

The proteins of the cellular plasma membrane (PM) perform important functions relating to homeostasis and intercellular communication. Due to its overall low cellular abundance, amphipathic character, and low membrane-to-cytoplasm ratio, the PM proteome has been challenging to isolate and characterize, and is poorly represented in standard LC-MS/MS analyses. In this study, we employ sucrose gradient ultracentrifugation for the enrichment of the PM proteome, without chemical labeling and affinity purification, together with GeLCMS and use subsequent bioinformatics tools to select proteins associated with the PM/cell surface, herein referred to as the surfaceome. Using this methodology, we identify over 1900 cell surface associated proteins in a human acute myeloid leukemia cell line. These surface proteins comprise almost 50% of all detected cellular proteins, a number that substantially exceeds the depth of coverage in previously published studies describing the leukemia surfaceome.


Subject(s)
Cell Membrane/chemistry , Computational Biology/methods , Leukocytes/chemistry , Membrane Proteins/isolation & purification , Cell Line, Tumor , Cell Membrane/metabolism , Centrifugation, Density Gradient , Chromatography, Liquid , Humans , Leukocytes/metabolism , Sucrose/chemistry , Tandem Mass Spectrometry , Ultracentrifugation/methods
13.
J Biol Chem ; 291(7): 3595-612, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26694611

ABSTRACT

The formation of functional amyloid must be carefully regulated to prevent the accumulation of potentially toxic products. Premelanosome protein (PMEL) forms non-toxic functional amyloid fibrils that assemble into sheets upon which melanins ultimately are deposited within the melanosomes of pigment cells. PMEL is synthesized in the endoplasmic reticulum but forms amyloid only within post-Golgi melanosome precursors; thus, PMEL must traverse the secretory pathway in a non-amyloid form. Here, we identified two pre-amyloid PMEL intermediates that likely regulate the timing of fibril formation. Analyses by non-reducing SDS-PAGE, size exclusion chromatography, and sedimentation velocity revealed two native high Mr disulfide-bonded species that contain Golgi-modified forms of PMEL. These species correspond to disulfide bond-containing dimeric and monomeric PMEL isoforms that contain no other proteins as judged by two-dimensional PAGE of metabolically labeled/immunoprecipitated PMEL and by mass spectrometry of affinity-purified complexes. Metabolic pulse-chase analyses, small molecule inhibitor treatments, and evaluation of site-directed mutants suggest that the PMEL dimer forms around the time of endoplasmic reticulum exit and is resolved by disulfide bond rearrangement into a monomeric form within the late Golgi or a post-Golgi compartment. Mutagenesis of individual cysteine residues within the non-amyloid cysteine-rich Kringle-like domain stabilizes the disulfide-bonded dimer and impairs fibril formation as determined by electron microscopy. Our data show that the Kringle-like domain facilitates the resolution of disulfide-bonded PMEL dimers and promotes PMEL functional amyloid formation, thereby suggesting that PMEL dimers must be resolved to monomers to generate functional amyloid fibrils.


Subject(s)
Amyloid/chemistry , Models, Molecular , Multivesicular Bodies/ultrastructure , Protein Processing, Post-Translational , gp100 Melanoma Antigen/chemistry , Amino Acid Substitution , Amyloid/metabolism , Amyloid/ultrastructure , Cell Line, Tumor , Cysteine/chemistry , Cysteine/metabolism , Cystine/chemistry , Cystine/metabolism , Dimerization , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kringles , Microscopy, Electron, Transmission , Molecular Weight , Multivesicular Bodies/chemistry , Multivesicular Bodies/metabolism , Mutagenesis, Site-Directed , Point Mutation , Protein Folding , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
14.
Mar Drugs ; 15(8)2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28783126

ABSTRACT

In previous work, we characterized the strong neuroprotective properties of the marine compound Psammaplysene A (PA) in in vitro and in vivo models of neurodegeneration. Based on its strong neuroprotective activity, the current work attempts to identify the physical target of PA to gain mechanistic insight into its molecular action. Two distinct methods, used in parallel, to purify protein-binding partners of PA led to the identification of HNRNPK as a direct target of PA. Based on surface plasmon resonance, we find that the binding of PA to HNRNPK is RNA-dependent. These findings suggest a role for HNRNPK-dependent processes in neurodegeneration/neuroprotection, and warrant further study of HNRNPK in this context.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Neuroprotective Agents/pharmacology , RNA-Binding Proteins/metabolism , Tyrosine/analogs & derivatives , HEK293 Cells , Humans , Marine Biology , Protein Binding , Tyrosine/pharmacology
15.
Am J Physiol Lung Cell Mol Physiol ; 311(6): L1062-L1075, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27694472

ABSTRACT

Alveolar epithelial regeneration is essential for resolution of the acute respiratory distress syndrome (ARDS). Although neutrophils have traditionally been considered mediators of epithelial damage, recent studies suggest they promote type II pneumocyte (AT2) proliferation, which is essential for regenerating alveolar epithelium. These studies did not, however, evaluate this relationship in an in vivo model of alveolar epithelial repair following injury. To determine whether neutrophils influence alveolar epithelial repair in vivo, we developed a unilateral acid injury model that creates a severe yet survivable injury with features similar to ARDS. Mice that received injections of the neutrophil-depleting Ly6G antibody had impaired AT2 proliferation 24 and 72 h after acid instillation, which was associated with decreased reepithelialization and increased alveolar protein concentration 72 h after injury. As neutrophil depletion itself may alter the cytokine response, we questioned the contribution of neutrophils to alveolar epithelial repair in neutropenic granulocyte-colony stimulating factor (G-CSF)-/- mice. We found that the loss of G-CSF recapitulated the neutrophil response of Ly6G-treated mice and was associated with defective alveolar epithelial repair, similar to neutrophil-depleted mice, and was reversed by administration of exogenous G-CSF. To approach the mechanisms, we employed an unbiased protein analysis of bronchoalveolar lavage fluid from neutrophil-depleted and neutrophil-replete mice 12 h after inducing lung injury. Pathway analysis identified significant differences in multiple signaling pathways that may explain the differences in epithelial repair. These data emphasize an important link between the innate immune response and tissue repair in which neutrophils promote alveolar epithelial regeneration.


Subject(s)
Acute Lung Injury/pathology , Alveolar Epithelial Cells/pathology , Epithelium/pathology , Neutrophils/pathology , Regeneration , Acids , Acute Lung Injury/chemically induced , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Antibodies/pharmacology , Bronchoalveolar Lavage Fluid , Cell Proliferation/drug effects , Disease Models, Animal , Epithelium/drug effects , Granulocyte Colony-Stimulating Factor/deficiency , Granulocyte Colony-Stimulating Factor/metabolism , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Proteomics , Regeneration/drug effects , Respiratory Distress Syndrome/pathology , Signal Transduction/drug effects , Up-Regulation/drug effects , Wound Healing/drug effects
16.
Nucleic Acids Res ; 42(8): 5347-60, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24567547

ABSTRACT

The stable ribonucleoprotein (RNP) complex formed between the Lactococcus lactis group II intron and its self-encoded LtrA protein is essential for the intron's genetic mobility. In this study, we report the biochemical, compositional, hydrodynamic and structural properties of active group II intron RNP particles (+A) isolated from its native host using a novel purification scheme. We employed small-angle X-ray scattering to determine the structural properties of these particles as they exist in solution. Using sucrose as a contrasting agent, we derived a two-phase quaternary model of the protein-RNA complex. This approach revealed that the spatial properties of the complex are largely defined by the RNA component, with the protein dimer located near the center of mass. A transfer RNA fusion engineered into domain II of the intron provided a distinct landmark consistent with this interpretation. Comparison of the derived +A RNP shape with that of the previously reported precursor intron (ΔA) particle extends previous findings that the loosely packed precursor RNP undergoes a dramatic conformational change as it compacts into its active form. Our results provide insights into the quaternary arrangement of these RNP complexes in solution, an important step to understanding the transition of the group II intron from the precursor to a species fully active for DNA invasion.


Subject(s)
Introns , Ribonucleoproteins/chemistry , Lactococcus lactis/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Multimerization , RNA, Transfer/chemistry , Ribonucleoproteins/isolation & purification , Ribonucleoproteins/metabolism , Scattering, Small Angle , X-Ray Diffraction
17.
Sci Adv ; 10(6): eadj4767, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38335281

ABSTRACT

Alpha-synuclein (αSyn) protein levels correlate with the risk and severity of Parkinson's disease and related neurodegenerative diseases. Lowering αSyn is being actively investigated as a therapeutic modality. Here, we systematically map the regulatory network that controls endogenous αSyn using sequential CRISPR-knockout and -interference screens in an αSyn gene (SNCA)-tagged cell line and induced pluripotent stem cell-derived neurons (iNeurons). We uncover αSyn modifiers at multiple regulatory layers, with amino-terminal acetyltransferase B (NatB) enzymes being the most potent endogenous αSyn modifiers in both cell lines. Amino-terminal acetylation protects the cytosolic αSyn from rapid degradation by the proteasome in a Ube2w-dependent manner. Moreover, we show that pharmacological inhibition of methionyl-aminopeptidase 2, a regulator of NatB complex formation, attenuates endogenous αSyn in iNeurons carrying SNCA triplication. Together, our study reveals several gene networks that control endogenous αSyn, identifies mechanisms mediating the degradation of nonacetylated αSyn, and illustrates potential therapeutic pathways for decreasing αSyn levels in synucleinopathies.


Subject(s)
N-Terminal Acetyltransferase B , Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , N-Terminal Acetyltransferase B/antagonists & inhibitors , N-Terminal Acetyltransferase B/metabolism , Methionyl Aminopeptidases/antagonists & inhibitors , Methionyl Aminopeptidases/metabolism
18.
J Thromb Haemost ; 22(5): 1447-1462, 2024 May.
Article in English | MEDLINE | ID: mdl-38160730

ABSTRACT

BACKGROUND: Recent clinical studies have shown that transfusions of adult platelets increase morbidity and mortality in preterm infants. Neonatal platelets are hyporesponsive to agonist stimulation, and emerging evidence suggests developmental differences in platelet immune functions. OBJECTIVES: This study was designed to compare the proteome and phosphoproteome of resting adult and neonatal platelets. METHODS: We isolated resting umbilical cord blood-derived platelets from healthy full-term neonates (n = 8) and resting blood platelets from healthy adults (n = 6) and compared protein and phosphoprotein contents using data-independent acquisition mass spectrometry. RESULTS: We identified 4770 platelet proteins with high confidence across all samples. Adult and neonatal platelets were clustered separately by principal component analysis. Adult platelets were significantly enriched in immunomodulatory proteins, including ß2 microglobulin and CXCL12, whereas neonatal platelets were enriched in ribosomal components and proteins involved in metabolic activities. Adult platelets were enriched in phosphorylated GTPase regulatory enzymes and proteins participating in trafficking, which may help prime them for activation and degranulation. Neonatal platelets were enriched in phosphorylated proteins involved in insulin growth factor signaling. CONCLUSION: Using label-free data-independent acquisition mass spectrometry, our findings expanded the known neonatal platelet proteome and identified important differences in protein content and phosphorylation between neonatal and adult platelets. These developmental differences suggested enhanced immune functions for adult platelets and presence of molecular machinery related to platelet activation. These findings are important to understanding mechanisms underlying key platelet functions as well as the harmful effects of adult platelet transfusions given to preterm infants.


Subject(s)
Blood Platelets , Fetal Blood , Phosphoproteins , Proteomics , Signal Transduction , Humans , Blood Platelets/metabolism , Infant, Newborn , Adult , Fetal Blood/metabolism , Fetal Blood/cytology , Phosphorylation , Proteomics/methods , Phosphoproteins/blood , Proteome , Female , Age Factors , Male , Principal Component Analysis , Mass Spectrometry , Tandem Mass Spectrometry
19.
J Neurosci ; 32(32): 11082-94, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875940

ABSTRACT

Anti-NMDA receptor (NMDAR) encephalitis is a newly identified autoimmune disorder that targets NMDARs, causing severe neurological symptoms including hallucinations, psychosis, and seizures, and may result in death (Dalmau et al., 2008). However, the exact epitope to which these antibodies bind is unknown. A clearly defined antigenic region could provide more precise testing, allow for comparison of immunogenicity between patients to explore potential clinically relevant variations, elucidate the functional effects of antibodies, and make patients' antibodies a more effective tool with which to study NMDAR function. Here, we use human CSF to explore the antigenic region of the NMDAR. We created a series of mutants within the amino terminal domain of GluN1 that change patient antibody binding in transfected cells in stereotyped ways. These mutants demonstrate that the N368/G369 region of GluN1 is crucial for the creation of immunoreactivity. Mass spectrometry experiments show that N368 is glycosylated in transfected cells and rat brain regions; however, this glycosylation is not directly required for epitope formation. Mutations of residues N368/G369 change the closed time of the receptor in single channel recordings; more frequent channel openings correlates with the degree of antibody staining, and acute antibody exposure prolongs open time of the receptor. The staining pattern of mutant receptors is similar across subgroups of patients, indicating consistent immunogenicity, although we have identified one region that has a variable role in epitope formation. These findings provide tools for detailed comparison of antibodies across patients and suggest an interaction between antibody binding and channel function.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/cerebrospinal fluid , Antibodies/cerebrospinal fluid , Epitopes/chemistry , Receptors, AMPA/chemistry , Receptors, N-Methyl-D-Aspartate/immunology , Age Factors , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/genetics , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/pathology , Antibodies/immunology , Binding Sites, Antibody/drug effects , Binding Sites, Antibody/genetics , Biotinylation , Brain/drug effects , Brain/metabolism , Cell Line, Transformed , Epitopes/metabolism , Female , Glutamic Acid/pharmacology , Glycine/pharmacology , Glycosylation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutagenesis, Site-Directed/methods , Mutation/genetics , Patch-Clamp Techniques , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Statistics, Nonparametric , Tandem Mass Spectrometry , Transfection , Tunicamycin/pharmacology
20.
J Biol Chem ; 287(46): 38449-59, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23012372

ABSTRACT

The HtrA protease of Streptococcus pneumoniae functions both in a general stress response role and as an error sensor that specifically represses genetic competence when the overall level of biosynthetic errors in cellular proteins is low. However, the mechanism through which HtrA inhibits development of competence has been unknown. We found that HtrA digested the pneumococcal competence-stimulating peptide (CSP) and constituted the primary extracytoplasmic CSP-degrading activity in cultures of S. pneumoniae. Mass spectrometry demonstrated that cleavage predominantly followed residue Phe-8 of the CSP-1 isoform of the peptide within its central hydrophobic patch, and in competition assays, both CSP-1 and CSP-2 interacted with HtrA with similar efficiencies. More generally, analysis of ß-casein digestion and of digestion within HtrA itself revealed a preference for substrates with non-polar residues at the P1 site. Consistent with a specificity for exposed hydrophobic residues, competition from native BSA only weakly inhibited digestion of CSP, but denaturation converted BSA into a strong competitive inhibitor of such proteolysis. Together these findings support a model in which digestion of CSP by HtrA is reduced in the presence of other unfolded proteins that serve as alternative targets for degradation. Such competition may provide a mechanism by which HtrA functions in a quality control capacity to monitor the frequency of biosynthetic errors that result in protein misfolding.


Subject(s)
Peptide Hydrolases/chemistry , Serine Proteases/physiology , Streptococcus pneumoniae/enzymology , Bacterial Physiological Phenomena , Binding Sites , Catalytic Domain , Fluorescence Resonance Energy Transfer , Gene Deletion , Kinetics , Mass Spectrometry/methods , Mutation , Peptides/chemistry , Polymerase Chain Reaction/methods , Protein Denaturation , Proteolysis , Quorum Sensing , Serine Proteases/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL